1
|
Jovanovic MZ, Stanojevic J, Stevanovic I, Ninkovic M, Ilic TV, Nedeljkovic N, Dragic M. Prolonged intermittent theta burst stimulation restores the balance between A2AR- and A1R-mediated adenosine signaling in the 6-hydroxidopamine model of Parkinson's disease. Neural Regen Res 2025; 20:2053-2067. [PMID: 39254566 PMCID: PMC11691459 DOI: 10.4103/nrr.nrr-d-23-01542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 04/30/2024] [Accepted: 06/17/2024] [Indexed: 09/11/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202507000-00027/figure1/v/2024-09-09T124005Z/r/image-tiff An imbalance in adenosine-mediated signaling, particularly the increased A2AR-mediated signaling, plays a role in the pathogenesis of Parkinson's disease. Existing therapeutic approaches fail to alter disease progression, demonstrating the need for novel approaches in PD. Repetitive transcranial magnetic stimulation is a non-invasive approach that has been shown to improve motor and non-motor symptoms of Parkinson's disease. However, the underlying mechanisms of the beneficial effects of repetitive transcranial magnetic stimulation remain unknown. The purpose of this study is to investigate the extent to which the beneficial effects of prolonged intermittent theta burst stimulation in the 6-hydroxydopamine model of experimental parkinsonism are based on modulation of adenosine-mediated signaling. Animals with unilateral 6-hydroxydopamine lesions underwent intermittent theta burst stimulation for 3 weeks and were tested for motor skills using the Rotarod test. Immunoblot, quantitative reverse transcription polymerase chain reaction, immunohistochemistry, and biochemical analysis of components of adenosine-mediated signaling were performed on the synaptosomal fraction of the lesioned caudate putamen. Prolonged intermittent theta burst stimulation improved motor symptoms in 6-hydroxydopamine-lesioned animals. A 6-hydroxydopamine lesion resulted in progressive loss of dopaminergic neurons in the caudate putamen. Treatment with intermittent theta burst stimulation began 7 days after the lesion, coinciding with the onset of motor symptoms. After treatment with prolonged intermittent theta burst stimulation, complete motor recovery was observed. This improvement was accompanied by downregulation of the eN/CD73-A2AR pathway and a return to physiological levels of A1R-adenosine deaminase 1 after 3 weeks of intermittent theta burst stimulation. Our results demonstrated that 6-hydroxydopamine-induced degeneration reduced the expression of A1R and elevated the expression of A2AR. Intermittent theta burst stimulation reversed these effects by restoring the abundances of A1R and A2AR to control levels. The shift in ARs expression likely restored the balance between dopamine-adenosine signaling, ultimately leading to the recovery of motor control.
Collapse
Affiliation(s)
- Milica Zeljkovic Jovanovic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Jelena Stanojevic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Ivana Stevanovic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Milica Ninkovic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Tihomir V. Ilic
- Medical Faculty of Military Medical Academy, University of Defence, Belgrade, Serbia
| | - Nadezda Nedeljkovic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| | - Milorad Dragic
- Laboratory for Neurobiology, Department of General Physiology and Biophysics, Faculty of Biology, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
2
|
Seplovich G, Bouchi Y, de Rivero Vaccari JP, Pareja JCM, Reisner A, Blackwell L, Mechref Y, Wang KK, Tyndall JA, Tharakan B, Kobeissy F. Inflammasome links traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Neural Regen Res 2025; 20:1644-1664. [PMID: 39104096 PMCID: PMC11688549 DOI: 10.4103/nrr.nrr-d-24-00107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/20/2024] [Accepted: 06/03/2024] [Indexed: 08/07/2024] Open
Abstract
Traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease are three distinct neurological disorders that share common pathophysiological mechanisms involving neuroinflammation. One sequela of neuroinflammation includes the pathologic hyperphosphorylation of tau protein, an endogenous microtubule-associated protein that protects the integrity of neuronal cytoskeletons. Tau hyperphosphorylation results in protein misfolding and subsequent accumulation of tau tangles forming neurotoxic aggregates. These misfolded proteins are characteristic of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease and can lead to downstream neuroinflammatory processes, including assembly and activation of the inflammasome complex. Inflammasomes refer to a family of multimeric protein units that, upon activation, release a cascade of signaling molecules resulting in caspase-induced cell death and inflammation mediated by the release of interleukin-1β cytokine. One specific inflammasome, the NOD-like receptor protein 3, has been proposed to be a key regulator of tau phosphorylation where it has been shown that prolonged NOD-like receptor protein 3 activation acts as a causal factor in pathological tau accumulation and spreading. This review begins by describing the epidemiology and pathophysiology of traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease. Next, we highlight neuroinflammation as an overriding theme and discuss the role of the NOD-like receptor protein 3 inflammasome in the formation of tau deposits and how such tauopathic entities spread throughout the brain. We then propose a novel framework linking traumatic brain injury, chronic traumatic encephalopathy, and Alzheimer's disease as inflammasome-dependent pathologies that exist along a temporal continuum. Finally, we discuss potential therapeutic targets that may intercept this pathway and ultimately minimize long-term neurological decline.
Collapse
Affiliation(s)
| | - Yazan Bouchi
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | - Juan Pablo de Rivero Vaccari
- Department of Neurological Surgery and the Miami Project to Cure Paralysis, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Jennifer C. Munoz Pareja
- Division of Pediatric Critical Care, Department of Pediatrics, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Andrew Reisner
- Department of Pediatrics, Emory University, Atlanta, GA, USA
- Department of Neurosurgery, Children’s Healthcare of Atlanta, Atlanta, GA, USA
| | - Laura Blackwell
- Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Yehia Mechref
- Department of Chemistry and Biochemistry, Texas Tech University, Lubbock, TX, USA
| | - Kevin K. Wang
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| | | | - Binu Tharakan
- Department of Surgery, Morehouse School of Medicine, Atlanta, GA, USA
| | - Firas Kobeissy
- Department of Neurobiology, Center for Neurotrauma, Multiomics & Biomarkers (CNMB), Neuroscience Institute, Morehouse School of Medicine, Atlanta, GA, USA
| |
Collapse
|
3
|
Cieri MB, Ramos AJ. Astrocytes, reactive astrogliosis, and glial scar formation in traumatic brain injury. Neural Regen Res 2025; 20:973-989. [PMID: 38989932 PMCID: PMC11438322 DOI: 10.4103/nrr.nrr-d-23-02091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 04/14/2024] [Indexed: 07/12/2024] Open
Abstract
Traumatic brain injury is a global health crisis, causing significant death and disability worldwide. Neuroinflammation that follows traumatic brain injury has serious consequences for neuronal survival and cognitive impairments, with astrocytes involved in this response. Following traumatic brain injury, astrocytes rapidly become reactive, and astrogliosis propagates from the injury core to distant brain regions. Homeostatic astroglial proteins are downregulated near the traumatic brain injury core, while pro-inflammatory astroglial genes are overexpressed. This altered gene expression is considered a pathological remodeling of astrocytes that produces serious consequences for neuronal survival and cognitive recovery. In addition, glial scar formed by reactive astrocytes is initially necessary to limit immune cell infiltration, but in the long term impedes axonal reconnection and functional recovery. Current therapeutic strategies for traumatic brain injury are focused on preventing acute complications. Statins, cannabinoids, progesterone, beta-blockers, and cerebrolysin demonstrate neuroprotective benefits but most of them have not been studied in the context of astrocytes. In this review, we discuss the cell signaling pathways activated in reactive astrocytes following traumatic brain injury and we discuss some of the potential new strategies aimed to modulate astroglial responses in traumatic brain injury, especially using cell-targeted strategies with miRNAs or lncRNA, viral vectors, and repurposed drugs.
Collapse
Affiliation(s)
- María Belén Cieri
- Laboratorio de Neuropatología Molecular, IBCN UBA-CONICET, Facultad de Medicina, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | |
Collapse
|
4
|
Pérez-Núñez R, González MF, Avalos AM, Leyton L. Impacts of PI3K/protein kinase B pathway activation in reactive astrocytes: from detrimental effects to protective functions. Neural Regen Res 2025; 20:1031-1041. [PMID: 38845231 PMCID: PMC11438337 DOI: 10.4103/nrr.nrr-d-23-01756] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 04/07/2024] [Accepted: 05/06/2024] [Indexed: 07/12/2024] Open
Abstract
Astrocytes are the most abundant type of glial cell in the central nervous system. Upon injury and inflammation, astrocytes become reactive and undergo morphological and functional changes. Depending on their phenotypic classification as A1 or A2, reactive astrocytes contribute to both neurotoxic and neuroprotective responses, respectively. However, this binary classification does not fully capture the diversity of astrocyte responses observed across different diseases and injuries. Transcriptomic analysis has revealed that reactive astrocytes have a complex landscape of gene expression profiles, which emphasizes the heterogeneous nature of their reactivity. Astrocytes actively participate in regulating central nervous system inflammation by interacting with microglia and other cell types, releasing cytokines, and influencing the immune response. The phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT) signaling pathway is a central player in astrocyte reactivity and impacts various aspects of astrocyte behavior, as evidenced by in silico , in vitro , and in vivo results. In astrocytes, inflammatory cues trigger a cascade of molecular events, where nuclear factor-κB serves as a central mediator of the pro-inflammatory responses. Here, we review the heterogeneity of reactive astrocytes and the molecular mechanisms underlying their activation. We highlight the involvement of various signaling pathways that regulate astrocyte reactivity, including the PI3K/AKT/mammalian target of rapamycin (mTOR), α v β 3 integrin/PI3K/AKT/connexin 43, and Notch/PI3K/AKT pathways. While targeting the inactivation of the PI3K/AKT cellular signaling pathway to control reactive astrocytes and prevent central nervous system damage, evidence suggests that activating this pathway could also yield beneficial outcomes. This dual function of the PI3K/AKT pathway underscores its complexity in astrocyte reactivity and brain function modulation. The review emphasizes the importance of employing astrocyte-exclusive models to understand their functions accurately and these models are essential for clarifying astrocyte behavior. The findings should then be validated using in vivo models to ensure real-life relevance. The review also highlights the significance of PI3K/AKT pathway modulation in preventing central nervous system damage, although further studies are required to fully comprehend its role due to varying factors such as different cell types, astrocyte responses to inflammation, and disease contexts. Specific strategies are clearly necessary to address these variables effectively.
Collapse
Affiliation(s)
- Ramón Pérez-Núñez
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - María Fernanda González
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Ana María Avalos
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Lisette Leyton
- Cellular Communication Laboratory, Programa de Biología Celular y Molecular, Center for Studies on Exercise, Metabolism and Cancer (CEMC), Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, Universidad de Chile, Santiago, Chile
| |
Collapse
|
5
|
Xie Q, Dasari R, Namba MD, Buck LA, Side CM, Park K, Jackson JG, Barker JM. Astrocytic regulation of cocaine locomotor sensitization in EcoHIV infected mice. Neuropharmacology 2025; 265:110245. [PMID: 39631679 DOI: 10.1016/j.neuropharm.2024.110245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/30/2024] [Accepted: 11/30/2024] [Indexed: 12/07/2024]
Abstract
Cocaine use disorder (CUD) is highly comorbid with HIV infection and worsens HIV outcomes. Preclinical research on the outcomes of HIV infection may yield crucial information on neurobehavioral changes resulting from chronic drug exposure in people living with HIV (PLWH). Repeated exposure to cocaine alters behavioral responses to cocaine. This includes development of cocaine locomotor sensitization - or increased locomotor responses to the same doses of cocaine - which depends on nucleus accumbens (NAc) neural plasticity. NAc astrocytes are key regulators of neural activity and plasticity, and their function can be impaired by cocaine exposure and HIV infection, thus implicating them as potential regulators of HIV-induced changes in behavioral response to cocaine. To characterize the effects of HIV infection on cocaine locomotor sensitization, we employed the EcoHIV mouse model in male and female mice to assess changes in locomotor responses after repeated cocaine (10 mg/kg) exposure and challenge. EcoHIV infection potentiated expression of cocaine sensitization. We also identified EcoHIV-induced increases in expression of the astrocytic nuclear marker Sox9 selectively in the NAc core. To investigate whether modulation of NAc astrocytes could reverse EcoHIV-induced deficits, we employed a chemogenetic approach. We found that chemogenetic activation of NAc astrocyte Gq signaling attenuated EcoHIV-enhanced cocaine sensitization. We propose that HIV infection contributes to cocaine behavioral sensitization and induces adaptations in NAc astrocytes, while promoting NAc astrocytic Gq-signaling can recover EcoHIV-induced behavioral changes. These findings identify potential cellular substrates of disordered cocaine-driven behavior in the context of HIV infection and point toward strategies to reduce cocaine-related behavior in PLWH.
Collapse
Affiliation(s)
- Qiaowei Xie
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA; Graduate Program in Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Rohan Dasari
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Mark D Namba
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Lauren A Buck
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Christine M Side
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Kyewon Park
- University of Pennsylvania Center for AIDS Research, USA
| | - Joshua G Jackson
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA
| | - Jacqueline M Barker
- Department of Pharmacology and Physiology, Drexel University College of Medicine, USA.
| |
Collapse
|
6
|
Jo MG, Hong J, Kim J, Kim SH, Lee B, Choi HN, Lee SE, Kim YJ, Park H, Park DH, Roh GS, Kim CS, Yun SP. Physiological change of striatum and ventral midbrain's glia cell in response to different exercise modalities. Behav Brain Res 2025; 479:115342. [PMID: 39571940 DOI: 10.1016/j.bbr.2024.115342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 11/06/2024] [Accepted: 11/15/2024] [Indexed: 11/27/2024]
Abstract
Exercise not only regulates neurotransmitters and synapse formation but also enhances the function of multiple brain regions, beyond cortical activation. Prolonged aerobic or resistance exercise modality has been widely applied to reveal the beneficial effects on the brain, but few studies have investigated the direct effects of different exercise modalities and variations in exercise intensity on the neuroinflammatory response in the brain and overall health. Therefore, in this study, we investigated changes in brain cells and the immune environment of the brain according to exercise modalities. This study was conducted to confirm whether different exercise modalities affect the location and function of dopaminergic neurons, which are responsible for regulating voluntary movement, before utilizing animal models of disease. The results showed that high-intensity interval exercise (HIE) increased the activity of A2-reactive astrocytes in the striatum (STR), which is directly involved in movement control, resulting in neuroprotective effects. Both HIE and combined exercises (CE) increased the expression of dopamine transporter (DAT) in the STR without damaging dopamine neurons in the ventral midbrain (VM). This means that exercise training can help improve and maintain exercise capacity. In conclusion, specific exercise modalities or intensity of exercise may contribute to preventing neurodegenerative diseases such as Parkinson's disease or enhancing therapeutic effects when combined with medication for patients with neurodegenerative diseases.
Collapse
Affiliation(s)
- Min Gi Jo
- Department of Pathology, College of Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Junyoung Hong
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
| | - Jiyeon Kim
- Institute of Sports & Arts Convergence (ISAC), Inha University, Incheon 22212, Republic of Korea
| | - Seon-Hee Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Bina Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Ha Nyeoung Choi
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - So Eun Lee
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Young Jin Kim
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Heejung Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea
| | - Dong-Ho Park
- Department of Biomedical Science, Program in Biomedical Science and Engineering, Inha University, Incheon 22212, Republic of Korea; Department of Kinesiology, Inha University, Incheon 22212, Republic of Korea
| | - Gu Seob Roh
- Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Anatomy, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea
| | - Chang Sun Kim
- Department of Physical Education, Dongduk Women's University, Seoul 02748, Republic of Korea.
| | - Seung Pil Yun
- Department of Pharmacology, Institute of Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea; Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju 52727, Republic of Korea.
| |
Collapse
|
7
|
Yu H, Ren K, Jin Y, Zhang L, Liu H, Huang Z, Zhang Z, Chen X, Yang Y, Wei Z. Mitochondrial DAMPs: Key mediators in neuroinflammation and neurodegenerative disease pathogenesis. Neuropharmacology 2025; 264:110217. [PMID: 39557152 DOI: 10.1016/j.neuropharm.2024.110217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/02/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Neurodegenerative diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), and amyotrophic lateral sclerosis (ALS) are increasingly linked to mitochondrial dysfunction and neuroinflammation. Central to this link are mitochondrial damage-associated molecular patterns (mtDAMPs), including mitochondrial DNA, ATP, and reactive oxygen species, released during mitochondrial stress or damage. These mtDAMPs activate inflammatory pathways, such as the NLRP3 inflammasome and cGAS-STING, contributing to the progression of neurodegenerative diseases. This review delves into the mechanisms by which mtDAMPs drive neuroinflammation and discusses potential therapeutic strategies targeting these pathways to mitigate neurodegeneration. Additionally, it explores the cross-talk between mitochondria and the immune system, highlighting the complex interplay that exacerbates neuronal damage. Understanding the role of mtDAMPs could pave the way for novel treatments aimed at modulating neuroinflammation and slowing disease progression, ultimately improving patient outcome.
Collapse
Affiliation(s)
- Haihan Yu
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Kaidi Ren
- Department of Pharmacy, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Yage Jin
- Department of Cardiology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Li Zhang
- Key Clinical Laboratory of Henan Province, Department of Clinical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China
| | - Hui Liu
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Zhen Huang
- Henan Key Laboratory of Immunology and Targeted Drug, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, School of Medical Technology, Xinxiang Medical University, Xinxiang, 453003, PR China
| | - Ziheng Zhang
- College of Life Sciences, Xinjiang University, Urumqi, Xinjiang, 830046, PR China
| | - Xing Chen
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Yang Yang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| | - Ziqing Wei
- Department of Neurology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, PR China.
| |
Collapse
|
8
|
Chen J, Zeng X, Wang L, Zhang W, Li G, Cheng X, Su P, Wan Y, Li X. Mutual regulation of microglia and astrocytes after Gas6 inhibits spinal cord injury. Neural Regen Res 2025; 20:557-573. [PMID: 38819067 PMCID: PMC11317951 DOI: 10.4103/nrr.nrr-d-23-01130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 12/05/2023] [Accepted: 01/17/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00032/figure1/v/2024-05-28T214302Z/r/image-tiff Invasive inflammation and excessive scar formation are the main reasons for the difficulty in repairing nervous tissue after spinal cord injury. Microglia and astrocytes play key roles in the spinal cord injury micro-environment and share a close interaction. However, the mechanisms involved remain unclear. In this study, we found that after spinal cord injury, resting microglia (M0) were polarized into pro-inflammatory phenotypes (MG1 and MG3), while resting astrocytes were polarized into reactive and scar-forming phenotypes. The expression of growth arrest-specific 6 (Gas6) and its receptor Axl were significantly down-regulated in microglia and astrocytes after spinal cord injury. In vitro experiments showed that Gas6 had negative effects on the polarization of reactive astrocytes and pro-inflammatory microglia, and even inhibited the cross-regulation between them. We further demonstrated that Gas6 can inhibit the polarization of reactive astrocytes by suppressing the activation of the Yes-associated protein signaling pathway. This, in turn, inhibited the polarization of pro-inflammatory microglia by suppressing the activation of the nuclear factor-κB/p65 and Janus kinase/signal transducer and activator of transcription signaling pathways. In vivo experiments showed that Gas6 inhibited the polarization of pro-inflammatory microglia and reactive astrocytes in the injured spinal cord, thereby promoting tissue repair and motor function recovery. Overall, Gas6 may play a role in the treatment of spinal cord injury. It can inhibit the inflammatory pathway of microglia and polarization of astrocytes, attenuate the interaction between microglia and astrocytes in the inflammatory microenvironment, and thereby alleviate local inflammation and reduce scar formation in the spinal cord.
Collapse
Affiliation(s)
- Jiewen Chen
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Xiaolin Zeng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Le Wang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Wenwu Zhang
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Gang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Xing Cheng
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Peiqiang Su
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Yong Wan
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| | - Xiang Li
- Department of Spine Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
- Guangdong Province Key Laboratory of Orthopedics and Traumatology, Guangzhou, Guangdong Province, China
| |
Collapse
|
9
|
Feng S, Li J, Liu T, Huang S, Chen X, Liu S, Zhou J, Zhao H, Hong Y. Overexpression of low-density lipoprotein receptor prevents neurotoxic polarization of astrocytes via inhibiting NLRP3 inflammasome activation in experimental ischemic stroke. Neural Regen Res 2025; 20:491-502. [PMID: 38819062 PMCID: PMC11317962 DOI: 10.4103/nrr.nrr-d-23-01263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/26/2023] [Accepted: 02/23/2024] [Indexed: 06/01/2024] Open
Abstract
JOURNAL/nrgr/04.03/01300535-202502000-00027/figure1/v/2024-05-28T214302Z/r/image-tiff Neurotoxic astrocytes are a promising therapeutic target for the attenuation of cerebral ischemia/reperfusion injury. Low-density lipoprotein receptor, a classic cholesterol regulatory receptor, has been found to inhibit NLR family pyrin domain containing protein 3 (NLRP3) inflammasome activation in neurons following ischemic stroke and to suppress the activation of microglia and astrocytes in individuals with Alzheimer's disease. However, little is known about the effects of low-density lipoprotein receptor on astrocytic activation in ischemic stroke. To address this issue in the present study, we examined the mechanisms by which low-density lipoprotein receptor regulates astrocytic polarization in ischemic stroke models. First, we examined low-density lipoprotein receptor expression in astrocytes via immunofluorescence staining and western blotting analysis. We observed significant downregulation of low-density lipoprotein receptor following middle cerebral artery occlusion reperfusion and oxygen-glucose deprivation/reoxygenation. Second, we induced the astrocyte-specific overexpression of low-density lipoprotein receptor using astrocyte-specific adeno-associated virus. Low-density lipoprotein receptor overexpression in astrocytes improved neurological outcomes in middle cerebral artery occlusion mice and reversed neurotoxic astrocytes to create a neuroprotective phenotype. Finally, we found that the overexpression of low-density lipoprotein receptor inhibited NLRP3 inflammasome activation in oxygen-glucose deprivation/reoxygenation injured astrocytes and that the addition of nigericin, an NLRP3 agonist, restored the neurotoxic astrocyte phenotype. These findings suggest that low-density lipoprotein receptor could inhibit the NLRP3-meidiated neurotoxic polarization of astrocytes and that increasing low-density lipoprotein receptor in astrocytes might represent a novel strategy for treating cerebral ischemic stroke.
Collapse
Affiliation(s)
- Shuai Feng
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Juanji Li
- Department of Neurology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu Province, China
| | - Tingting Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shiqi Huang
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Xiangliang Chen
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Shen Liu
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Junshan Zhou
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Hongdong Zhao
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| | - Ye Hong
- Department of Neurology, Nanjing First Hospital, Nanjing Medical University, Nanjing, Jiangsu Province, China
| |
Collapse
|
10
|
Absalyamova M, Traktirov D, Burdinskaya V, Artemova V, Muruzheva Z, Karpenko M. Molecular basis of the development of Parkinson's disease. Neuroscience 2025; 565:292-300. [PMID: 39653246 DOI: 10.1016/j.neuroscience.2024.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 12/13/2024]
Abstract
Parkinson's disease is one of the most prevalent neurodegenerative motor disorders worldwide with postural instability, bradykinesia, resting tremor and rigidity being the most common symptoms of the disease. Despite the fact that the molecular mechanisms of Parkinson's disease pathogenesis have already been well described, there is still no coherent picture of the etiopathogenesis of this disease. According to modern concepts, neurodegeneration is induced mainly by oxidative stress, neuroinflammation, dysregulation of cerebral proteostasis, apoptotic dysregulation, and impaired autophagy. This review describes how various factors contribute to neurodegeneration in Parkinson's disease. Understanding the factors affecting fundamental cellular processes and responsible for disease progression may help develop therapeutic strategies to improve the quality of life of patients suffering from the disease. The review also discusses the role of calpains in the development of Parkinson's disease. It is known that α-synuclein is a substrate of calcium-dependent proteases of the calpain family. Truncated forms of α-synuclein are not only involved in the process of formation of the aggregates, but also increase their toxicity.
Collapse
Affiliation(s)
| | | | | | | | | | - Marina Karpenko
- Peter the Great St Petersburg Polytechnic University, Russia
| |
Collapse
|
11
|
Carvalho D, Diaz-Amarilla P, Smith MR, Santi MD, Martinez-Busi M, Go YM, Jones DP, Duarte P, Savio E, Abin-Carriquiry JA, Arredondo F. Untargeted metabolomics of 3xTg-AD neurotoxic astrocytes. J Proteomics 2025; 310:105336. [PMID: 39448026 DOI: 10.1016/j.jprot.2024.105336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 10/17/2024] [Accepted: 10/19/2024] [Indexed: 10/26/2024]
Abstract
Alzheimer's disease (AD) is the most common form of dementia, affecting approximately 47 M people worldwide. Histological features and genetic risk factors, among other evidence, supported the amyloid hypothesis of the disease. This neuronocentric paradigm is currently undergoing a shift, considering evidence of the role of other cell types, such as microglia and astrocytes, in disease progression. Previously, we described a particular astrocyte subtype obtained from the 3xTg-AD murine model that displays neurotoxic properties in vitro. We continue here our exploratory analysis through the lens of metabolomics to identify potentially altered metabolites and biological pathways. Cell extracts from neurotoxic and control astrocytes were compared using high-resolution mass spectrometry-based metabolomics. Around 12 % of metabolic features demonstrated significant differences between neurotoxic and control astrocytes, including alterations in the key metabolite glutamate. Consistent with our previous transcriptomic study, the present results illustrate many homeostatic and regulatory functions of metabolites, suggesting that neurotoxic 3xTg-AD astrocytes exhibit alterations in the Krebs cycle as well as the prostaglandin pathway. This is the first metabolomic study performed in 3xTg-AD neurotoxic astrocytes. These results provide insight into metabolic alterations potentially associated with neurotoxicity and pathology progression in the 3xTg-AD mouse model and strengthen the therapeutic potential of astrocytes in AD. BIOLOGICAL SIGNIFICANCE: Our study is the first high-resolution metabolomic characterization of the novel neurotoxic 3xTg-AD astrocytes. We propose key metabolites and pathway alterations, as well as possible associations with gene expression alterations in the model. Our results are in line with recent hypotheses beyond the amyloid cascade, considering the involvement of several stress response cascades during the development of Alzheimer's disease. This work could inspire other researchers to initiate similar studies in related models. Furthermore, this work illustrates a powerful workflow for metabolite annotation and selection that can be implemented in other studies.
Collapse
Affiliation(s)
- Diego Carvalho
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay; Área de Matemática - DETEMA, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Pablo Diaz-Amarilla
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Mathew R Smith
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine; Department of Medicine, Emory University, GA, USA; Atlanta Veterans Affairs Healthcare System, Decatur, GA, USA
| | - María Daniela Santi
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay; Instituto Multidisciplinario de Biología Vegetal (IMBIV-CONICET), Ciudad Universitaria. X5000HUA, Córdoba, Argentina
| | - Marcela Martinez-Busi
- Plataforma de Servicios Analíticos, Instituto de Investigaciones Biológicas Clemente Estable, Montevideo, Uruguay
| | - Young-Mi Go
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine; Department of Medicine, Emory University, GA, USA
| | - Dean P Jones
- Division of Pulmonary, Allergy, Critical Care and Sleep Medicine; Department of Medicine, Emory University, GA, USA
| | - Pablo Duarte
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Eduardo Savio
- I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay
| | - Juan A Abin-Carriquiry
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay; Laboratorio de Biofármacos, Instituto Pasteur de Montevideo, Montevideo, Uruguay.
| | - Florencia Arredondo
- Departamento de Neuroquímica, Instituto de Investigaciones Biológicas Clemente Estable (IIBCE), Montevideo, Uruguay; I&D Biomédico y Químico Farmacéutico, Centro Uruguayo de Imagenología Molecular (CUDIM), Montevideo, Uruguay.
| |
Collapse
|
12
|
Wu W, Zhao Y, Cheng X, Xie X, Zeng Y, Tao Q, Yang Y, Xiao C, Zhang Z, Pang J, Jin J, He H, Lin Y, Li B, Ma J, Ye X, Lin WJ. Modulation of glymphatic system by visual circuit activation alleviates memory impairment and apathy in a mouse model of Alzheimer's disease. Nat Commun 2025; 16:63. [PMID: 39747869 PMCID: PMC11696061 DOI: 10.1038/s41467-024-55678-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Accepted: 12/20/2024] [Indexed: 01/04/2025] Open
Abstract
Alzheimer's disease is characterized by progressive amyloid deposition and cognitive decline, yet the pathological mechanisms and treatments remain elusive. Here we report the therapeutic potential of low-intensity 40 hertz blue light exposure in a 5xFAD mouse model of Alzheimer's disease. Our findings reveal that light treatment prevents memory decline in 4-month-old 5xFAD mice and motivation loss in 14-month-old 5xFAD mice, accompanied by restoration of glial water channel aquaporin-4 polarity, improved brain drainage efficiency, and a reduction in hippocampal lipid accumulation. We further demonstrate the beneficial effects of 40 hertz blue light are mediated through the activation of the vLGN/IGL-Re visual circuit. Notably, concomitant use of anti-Aβ antibody with 40 hertz blue light demonstrates improved soluble Aβ clearance and cognitive performance in 5xFAD mice. These findings offer functional evidence on the therapeutic effects of 40 hertz blue light in Aβ-related pathologies and suggest its potential as a supplementary strategy to augment the efficacy of antibody-based therapy.
Collapse
Affiliation(s)
- Wen Wu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yubai Zhao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
- Department of Clinical and Rehabilitation Medicine, Guiyang Healthcare Vocational University, Guizhou, China
| | - Xin Cheng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Xiaoru Xie
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Yixiu Zeng
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Quan Tao
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Yishuai Yang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
| | - Chuan Xiao
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Zhan Zhang
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China
| | - Jiahui Pang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Jin
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Hongbo He
- Guangdong Mental Health Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yangyang Lin
- Department of Rehabilitation Medicine, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Clinical Research Center for Rehabilitation Medicine, Guangzhou, China
- Biomedical Innovation Center, the Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Boxing Li
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China
- Advanced Medical Technology Center, the First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-Sen University), Ministry of Education, Guangzhou, China
| | - Junxian Ma
- Tianfu Xinglong Lake Laboratory, Chengdu, China.
| | - Xiaojing Ye
- Faculty of Forensic Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Guangdong Province Translational Forensic Medicine Engineering Technology Research Center, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
| | - Wei-Jye Lin
- Guangdong Provincial Key Laboratory of Brain Function and Disease, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, China.
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan, China.
| |
Collapse
|
13
|
Castro MML, Amaral Junior FLD, Mendes FDCCDS, Anthony DC, Brites DMTDO, Diniz CWP, Sosthenes MCK. Intriguing astrocyte responses in CA1 to reduced and rehabilitated masticatory function: Dorsal and ventral distinct perspectives in adult mice. Arch Oral Biol 2025; 169:106097. [PMID: 39395318 DOI: 10.1016/j.archoralbio.2024.106097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 09/05/2024] [Accepted: 09/26/2024] [Indexed: 10/14/2024]
Abstract
OBJECTIVE We sought to investigate the plasticity of diet-induced changes in astrocyte morphology of stratum lacunosum-moleculare (SLM) in CA1. DESIGN Three diet regimes were adopted in 15 mice, from the 21st postnatal day to 6 months. The first diet regimen was pellet feed, called Hard Diet (HD). The second, with reduced masticatory, received a pellet-diet followed by a powdered-diet, and it was identified as Hard Diet/Soft Diet (HD/SD). Finally, the group with rehabilitated masticatory was named Hard Diet/Soft Diet/Hard Diet (HD/SD/HD). In the end, euthanasia and brain histological processing were performed, in which astrocytic immunoreactivity to glial-fibrillary-acidic-protein (GFAP) was tested. In reconstructed astrocytes, morphometric analysis was performed. RESULTS Astrocyte morphometric revealed that changes in masticatory regimens impact astrocyte morphology. In the dorsal CA1, switching from a hard diet to a soft diet led to reductions in most variables, whereas in the ventral, fewer variables were affected, highlighting regional differences in astrocyte responses. Cluster analysis further showed that diet-induced changes in astrocyte morphology were reversible in the dorsal region, but not in the ventral region, indicating a persistent impact on astrocyte diversity and complexity in the ventral even after rehabilitation. Correlation tests between astrocyte morphology and behavioral performance demonstrated disrupted relationships under masticatory stress, with effects persisting after rehabilitation. CONCLUSION Changes in the diet result in significant alterations in astrocyte morphology, suggesting a direct link between dietary modulation and cellular structure. Morphometric analyses revealed distinct alterations in astrocyte morphology in response to changes in the masticatory regimen, with both dorsal/ventral regions displaying notable changes. Moreover, the regional differential effects on astrocytes underscore the complexity of mastication on neuroplasticity and cognitive function.
Collapse
Affiliation(s)
- Micaele Maria Lopes Castro
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA 66073-005, Brazil
| | - Fabio Leite do Amaral Junior
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA 66073-005, Brazil
| | - Fabíola de Carvalho Chaves de Siqueira Mendes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA 66073-005, Brazil; Curso de Medicina, Centro Universitário do Estado do Pará, Belém, PA 66613-903, Brazil
| | - Daniel Clive Anthony
- University of Oxford, Laboratory of Experimental Neuropathology, Department of Pharmacology, Oxford OX13QT, United Kingdom
| | - Dora Maria Tuna de Oliveira Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal; Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, Lisbon, Portugal
| | - Cristovam Wanderley Picanço Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA 66073-005, Brazil
| | - Marcia Consentino Kronka Sosthenes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém, PA 66073-005, Brazil.
| |
Collapse
|
14
|
Tsering W, de la Rosa A, Ruan IY, Philips JL, Bathe T, Villareal JA, Prokop S. Preferential clustering of microglia and astrocytes around neuritic plaques during progression of Alzheimer's disease neuropathological changes. J Neurochem 2025; 169:e16275. [PMID: 39655787 PMCID: PMC11629606 DOI: 10.1111/jnc.16275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/28/2024] [Accepted: 11/17/2024] [Indexed: 12/13/2024]
Abstract
Neuroinflammation plays an important role in the pathological cascade of Alzheimer's disease (AD) along with aggregation of extracellular amyloid-β (Aβ) plaques and intracellular aggregates of tau protein. In animal models of amyloidosis, local immune activation is centered around Aβ plaques, which are usually of uniform morphology, dependent on the transgenic model used. In postmortem human brains a diversity of Aβ plaque morphologies is seen including diffuse plaques (non-neuritic plaques, non-NP), dense-core plaques, cotton-wool plaques, and NP. In a recent study, we demonstrated that during the progression of Alzheimer's disease neuropathologic changes (ADNC), a transformation of non-NP into NP occurs which is tightly linked to the emergence of cortical, but not hippocampal neurofibrillary tangle (NFT) pathology. This highlights the central role of NP in AD pathogenesis as well as brain region-specific differences in NP formation. In order to correlate the transformation of plaque types with local immune activation, we quantified the clustering and phenotype of microglia and accumulation of astrocytes around non-NP and NP during the progression of ADNC. We hypothesize that glial clustering occurs in response to formation of neuritic dystrophy around NP. First, we show that Iba1-positive microglia preferentially cluster around NP. Utilizing microglia phenotypic markers, we furthermore demonstrate that CD68-positive phagocytic microglia show a strong preference to cluster around NP in both the hippocampus and frontal cortex. A similar preferential clustering is observed for CD11c and ferritin-positive microglia in the frontal cortex, while this preference is less pronounced in the hippocampus, highlighting differences between hippocampal and cortical Aβ plaques. Glial fibrillary acidic protein-positive astrocytes showed a clear preference for clustering around NP in both the frontal cortex and hippocampus. These data support the notion that NP are intimately associated with the neuroimmune response in AD and underscore the importance of the interplay of protein deposits and the immune system in the pathophysiology of AD.
Collapse
Affiliation(s)
- Wangchen Tsering
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Neuroscience, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- McKnight Brain Institute, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Ana de la Rosa
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Isabelle Y. Ruan
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Jennifer L. Philips
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Tim Bathe
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Jonathan A. Villareal
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
| | - Stefan Prokop
- Center for Translational Research in Neurodegenerative Disease, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- McKnight Brain Institute, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Department of Pathology, College of MedicineUniversity of FloridaGainesvilleFloridaUSA
- Norman Fixel Institute for Neurological DiseasesUniversity of FloridaGainesvilleFloridaUSA
| |
Collapse
|
15
|
Sommer S, Panzer A, Bertolini A, Cleaveland R, Jain V, Kapanci T, Derichs U, Geis T, Neu A, Löhr-Nilles C, Aeschimann-Huhn R, Flotats-Bastardas M, Deiva K, Armangue T, Olivé-Cirera G, Kannoth S, Koy A, Meirson H, Fattal-Valevski A, Ganelin-Cohen E, Losch H, Horne A, Wickström R, Dargvainiene J, Leypoldt F, Rostasy K. Spectrum of Clinical and Imaging Features of Children With GFAP Astrocytopathy. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2025; 12:e200327. [PMID: 39566024 DOI: 10.1212/nxi.0000000000200327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/03/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND AND OBJECTIVES Glial fibrillary acidic protein (GFAP) antibodies (abs) have been described primarily in adults with a spectrum of autoimmune-mediated diseases. In children, data on clinical and neuroradiologic features of children with autoimmune GFAP astrocytopathy are limited. The aim of this study was to describe the clinical and radiologic features in children with GFAP-ab-associated diseases. METHODS We retrospectively recruited children from 13 clinical centers between 2020 and 2023 who (1) tested positive for GFAP-ab in serum and/or CSF and (2) of whom a complete clinical and MRI data set was available. RESULTS We identified and included 15 children (5 girls, 10 boys). The median age at onset was 9.9 years (range: 2-16 years). All children presented with features of AE or meningitis, acute cerebellitis, or transverse myelitis. CSF pleocytosis was common (13/15, median 245 cells/μL), and 13 (87%) of 15 harbored GFAP-abs in their CSF, 8 (53%) of whom did not have detectable GFAP-abs in their serum. MRI was abnormal in 15 (100%) of 15 children: Specific patterns included confluent lesions in the pons or caudate nucleus (11/15; 73%), peri-aqueductal regions (13/15; 87%), and spinal cord (6/10; 60%). 12 children had a favorable outcome (mRS score of = 1). Two patients died in the acute phase or during follow-up. DISCUSSION GFAP-ab-associated diseases encompass a wide spectrum of clinical presentation associated with a particular set of MRI features clearly distinct to other antibody-mediated diseases or MOGAD. We recommend that testing for GFAP-abs in serum and CSF be included in the workup of children with AE, particularly if brainstem involvement occurs.
Collapse
Affiliation(s)
- Simon Sommer
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Andreas Panzer
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Annikki Bertolini
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Robert Cleaveland
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Vivek Jain
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Tugba Kapanci
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Ute Derichs
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Tobias Geis
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Axel Neu
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Christa Löhr-Nilles
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Rahel Aeschimann-Huhn
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Marina Flotats-Bastardas
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Kumaran Deiva
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Thais Armangue
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Gemma Olivé-Cirera
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Sudheeran Kannoth
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Anne Koy
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Hadas Meirson
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Aviva Fattal-Valevski
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Esther Ganelin-Cohen
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Heike Losch
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Annacarin Horne
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Ronny Wickström
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Justina Dargvainiene
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Frank Leypoldt
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| | - Kevin Rostasy
- From the Departments of Pediatric Neurology (S.S., A.B., K.R.), and Pediatric Radiology (A.P., R.C.), Children's Hospital Datteln, Witten/Herdecke University, Datteln, Germany; Consultant Child Neurologist and Epileptologist at Neoclinic Children's Hospital (V.J.), Jaipur, India; Department of Pediatric Neurology (T.K.), Children's Hospital Datteln, University Witten/Herdecke; Faculty of Health (T.K.), Department of Psychology and Psychotherapy, Chair of Personality Psychology and Diagnosis, Witten/Herdecke University; Center for Paediatric and Adolescent Medicine (U.D.), University Medical Clinic, Mainz; University Children's Hospital Regensburg (KUNO) (T.G.), Hospital St. Hedwig of the Order of St. John, University of Regensburg; Department of Pediatric Neurology (A.N.), VAMED Klinik Geesthacht; Department of Pediatrics (A.N.), University Medical Center Hamburg-Eppendorf; Department of Pediatric Neurology (C.L.-N.), Mutterhaus der Borromäerinnen, Trier; Department of Pediatric Intensive Care (R.A.-H.), University Children's Hal Marburg; Department of Pediatric Neurology (M.F.-B.), Saarland University Medical Center, Homburg/Saar, Germany; Assistance Publique-Hôpitaux de Paris (K.D.), Paris-Saclay University Hospitals, Bicêtre Hospital, Pediatric Neurology Department, National Referral Center for Rare Inflammatory and Auto-immune Brain and Spinal Diseases, Paris Saclay University, France; Neuroimmunology Unit (T.A.), in Sant Joan de Déu Children's Hospital, Esplugues de Llobregat, Barcelona; Neuroimmunology Program (T.A., G.O.-C.), Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Hospital Clínic, University of Barcelona; Neurology Unit (G.O.-C.), Hospital Parc Taulí de Sabadell, Sabadell, Barcelona, Spain; Neuroimmunology Laboratory (S.K.), Amrita Institute of Medical Sciences, School of Medicine, Amrita University, Kochi, India; Department of Pediatrics (A.K.); Center for Rare Diseases (A.K.), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Department of Pediatric Neurology (H.M.); Pediatric Neurology Institute (A.F.-V.), Dana-Dwek Children's Hospital, Tel Aviv Sourasky Medical Center; Sackler Faculty of Medicine, Tel Aviv University; Institute of Pediatric Neurology (E.G.-C.), Schneider Children's Medical Center of Israel, Petah Tikva, Israel; Sackler Faculty of Medicine, Tel Aviv University, Israel; University Children's Hospital Oldenburg (H.L.), Department of Neuropediatrics, Oldenburg; Neuropediatric Unit (A.H., R.W.), Karolinska University Hospital and Karolinska Institute Stockholm, Sweden; and Institute of Clinical Chemistry (J.D., F.L.), Neuroimmunology Unit and Department of Neurology, University Medical Center Schleswig-Holstein Campus, Kiel, Germany
| |
Collapse
|
16
|
Seady M, Schirmbeck G, Taday J, Fróes FT, Baú JV, Jantsch J, Guedes RP, Gonçalves CA, Leite MC. Curcumin attenuates neuroinflammatory damage induced by LPS: Implications for the role of S100B. J Nutr Biochem 2025; 135:109768. [PMID: 39278425 DOI: 10.1016/j.jnutbio.2024.109768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/24/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Inflammation is a common feature of neurological disorders that alters cell function in microglia and astrocytes as well as other neuronal cell types. Astrocytes modulate blood flow, regulate glutamate metabolism, and exert antioxidant protection. When responding to inflammatory damage, astrocytes enhance immune cell infiltration and amplify inflammatory responses via the upregulation of cytokine production. Several molecules have been proposed to attenuate neuroinflammation and control neurological diseases. Curcumin gained attention due to its capacity to cross the blood-brain barrier and its well-described anti-inflammatory and antioxidant activities. Our study aimed to understand if oral curcumin administration could protect against central nervous system inflammatory damage induced by intracerebroventricular injection of LPS while focusing on astrocyte function. Despite its poor bioavailability, we found that curcumin reaches the central nervous system, prevents the locomotory damage caused by LPS, and reduces inflammatory signaling via IL-1β and COX-2. Furthermore, we observed that curcumin was protective against LPS-induced S100B secretion in the cerebrospinal fluid and GSH reduction in the hippocampal tissue. However, curcumin could not protect the animals from anhedonia, assessed by the sucrose preference test, and weight loss induced by LPS. Our results indicate that oral curcumin administration exerts a protective anti-inflammatory action in the central nervous system, attenuating the sickness behavior induced by ICV LPS. This work demonstrates that curcumin has an important modulative effect on astrocytes, thus suggesting that astrocytes are critical to the anti-inflammatory effects of curcumin.
Collapse
Affiliation(s)
- Marina Seady
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriel Schirmbeck
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéssica Taday
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Telles Fróes
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jéfeli Vasques Baú
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Carlos-Alberto Gonçalves
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Marina Concli Leite
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| |
Collapse
|
17
|
Cabral-Miranda F, Matias I, Gomes FCA. Astrocytic proteostasis in the tale of aging and neurodegeneration. Ageing Res Rev 2025; 103:102580. [PMID: 39557299 DOI: 10.1016/j.arr.2024.102580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/05/2024] [Accepted: 11/09/2024] [Indexed: 11/20/2024]
Abstract
Homeostasis of proteins (proteostasis), which governs protein processing, folding, quality control, and degradation, is a fundamental cellular process that plays a pivotal role in various neurodegenerative diseases and in the natural aging process of the mammalian brain. While the role of neuronal proteostasis in neuronal physiology is well characterized, the contribution of proteostasis of glial cells, particularly of astrocytes, has received fairly less attention in this context. Here, we summarize recent data highlighting proteostasis dysfunction in astrocytes and its putative implication to neurodegenerative diseases and aging. We discuss how distinct proteostasis nodes and pathways in astrocytes may specifically contribute to brain function and different age-associated pathologies. Finally, we argue that the understanding of astrocytic proteostasis role in neuronal physiology and functional decay may arise as a potential new avenue of intervention in neurodegenerative diseases and grant relevant data in the biology of aging.
Collapse
Affiliation(s)
- Felipe Cabral-Miranda
- Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Isadora Matias
- Institute of Biomedical Sciences, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
18
|
Demmings MD, da Silva Chagas L, Traetta ME, Rodrigues RS, Acutain MF, Barykin E, Datusalia AK, German-Castelan L, Mattera VS, Mazengenya P, Skoug C, Umemori H. (Re)building the nervous system: A review of neuron-glia interactions from development to disease. J Neurochem 2025; 169:e16258. [PMID: 39680483 DOI: 10.1111/jnc.16258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 12/18/2024]
Abstract
Neuron-glia interactions are fundamental to the development and function of the nervous system. During development, glia, including astrocytes, microglia, and oligodendrocytes, influence neuronal differentiation and migration, synapse formation and refinement, and myelination. In the mature brain, glia are crucial for maintaining neural homeostasis, modulating synaptic activity, and supporting metabolic functions. Neurons, inherently vulnerable to various stressors, rely on glia for protection and repair. However, glia, in their reactive state, can also promote neuronal damage, which contributes to neurodegenerative and neuropsychiatric diseases. Understanding the dual role of glia-as both protectors and potential aggressors-sheds light on their complex contributions to disease etiology and pathology. By appropriately modulating glial activity, it may be possible to mitigate neurodegeneration and restore neuronal function. In this review, which originated from the International Society for Neurochemistry (ISN) Advanced School in 2019 held in Montreal, Canada, we first describe the critical importance of glia in the development and maintenance of a healthy nervous system as well as their contributions to neuronal damage and neurological disorders. We then discuss potential strategies to modulate glial activity during disease to protect and promote a properly functioning nervous system. We propose that targeting glial cells presents a promising therapeutic avenue for rebuilding the nervous system.
Collapse
Affiliation(s)
- Matthew D Demmings
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Luana da Silva Chagas
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Rio de Janeiro, Brazil
| | - Marianela E Traetta
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Rui S Rodrigues
- University of Bordeaux, INSERM, Neurocentre Magendie U1215, Bordeaux, France
| | - Maria Florencia Acutain
- Instituto de Biología Celular y Neurociencia (IBCN), Facultad de Medicina, Conicet, Buenos Aires, Argentina
| | - Evgeny Barykin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Ashok Kumar Datusalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER Raebareli), Raebareli, UP, India
| | - Liliana German-Castelan
- Neuroscience Program, Schulich School of Medicine and Dentistry, University of Western Ontario, London, Ontario, Canada
| | - Vanesa S Mattera
- Instituto de Química y Fisicoquímica Biológica (IQUIFIB-FFyB-UBA), Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Pedzisai Mazengenya
- Center of Medical and bio-Allied Health Sciences Research, College of Medicine, Ajman University, Ajman, United Arab Emirates
| | - Cecilia Skoug
- Department of Neuroscience, Physiology & Pharmacology, Centre for Cardiovascular and Metabolic Neuroscience, University College London, London, UK
| | - Hisashi Umemori
- Department of Neurology, F.M. Kirby Neurobiology Center, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
19
|
Sampson E, Mills NT, Hori H, Cearns M, Schwarte K, Hohoff C, Oliver Schubert K, Fourrier C, Baune BT. Long-term characterisation of the relationship between change in depression severity and change in inflammatory markers following inflammation-stratified treatment with vortioxetine augmented with celecoxib or placebo. Brain Behav Immun 2025; 123:43-56. [PMID: 39243988 DOI: 10.1016/j.bbi.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/08/2024] [Accepted: 09/03/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Major depressive disorder (MDD) is a highly prevalent condition with a substantial incidence of relapse or treatment resistance. A subset of patients show evidence of low-grade inflammation, with these patients having a higher likelihood of more severe or difficult to treat courses of illness. Anti-inflammatory treatment of MDD has been investigated with mixed results, and no known studies have included assessments beyond cessation of the anti-inflammatory agent, meaning it remains unknown if any benefit from treatment persists. The objective of the present study was to investigate treatment outcomes up to 29 weeks post-cessation of celecoxib or placebo augmentation of an antidepressant, and how concentrations of selected inflammatory markers change over the same period. METHODS The PREDDICT parallel-group, randomised, double-blind, placebo-controlled trial (University of Adelaide, Australia) ran from December 2017 to April 2020. Participants with MDD were stratified into normal range or elevated inflammation strata according to screening concentrations of high sensitivity C-reactive protein (hsCRP). Participants were randomised to treatment with vortioxetine and celecoxib or vortioxetine and placebo for six weeks, and vortioxetine alone for an additional 29 weeks (35 total weeks). Following a previous publication of results from the six-week RCT phase, exploratory analyses were performed on Montgomery-Åsberg Depression Rating Scale (MADRS) scores, response and remission outcomes, and selected peripheral inflammatory markers across the entire study duration up to week 35. RESULTS Participants retained at each observation were baseline N=119, week 2 N=115, week 4 N=103, week 6 N=104, week 8 N=98, week 22 N=81, and week 35 N=60. Those in the elevated hsCRP celecoxib-augmented group had a statistically significantly greater reduction in MADRS score from baseline to week 35 compared to all other groups, demonstrating the greatest clinical improvement long-term, despite no group or strata differences at preceding time points. Response and remission outcomes did not differ by treatment group or hsCRP strata at any time point. Changes in hsCRP between baseline and week 35 and Tumour Necrosis Factor-α (TNF-α) concentrations between baseline and week 6 and baseline and week 35 were statistically significantly associated with MADRS scores observed at week 6 and week 35 respectively, with reducing TNF-α concentrations associated with reducing MADRS scores and vice versa in each case. A post-hoc stratification of the participant cohort by baseline TNF-α concentrations led to significant prediction by the derived strata on clinical response at weeks 6, 8 and 35, with participants with elevated baseline TNF-α less likely to achieve clinical response. INTERPRETATION The present analysis suggests for the first time a possible longer-term clinical benefit of celecoxib augmentation of vortioxetine in inflammation-associated MDD treatment. However, further research is needed to confirm the finding and to ascertain the reason for such a delayed effect. Furthermore, the trial suggests that TNF-α may have a stronger relationship with anti-inflammatory MDD treatment outcomes than hsCRP, and should be investigated further for potential predictive utility. CLINICAL TRIALS REGISTRATION Australian New Zealand Clinical Trials Registry (ANZCTR), ACTRN12617000527369p. Registered on 11 April 2017, http://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?ACTRN=12617000527369p.
Collapse
Affiliation(s)
- Emma Sampson
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Natalie T Mills
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Hikaru Hori
- Department of Psychiatry, Faculty of Medicine, Fukuoka University, Fukuoka City, Japan
| | - Micah Cearns
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Kathrin Schwarte
- Department of Psychiatry, University of Münster, Münster, Germany
| | - Christa Hohoff
- Department of Psychiatry, University of Münster, Münster, Germany
| | - K Oliver Schubert
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Northern Adelaide Mental Health Service, Salisbury, Australia
| | - Célia Fourrier
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Bernhard T Baune
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, Australia; Department of Psychiatry, University of Münster, Münster, Germany; Department of Psychiatry, Melbourne Medical School, The University of Melbourne, Melbourne, Australia; The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia.
| |
Collapse
|
20
|
Sasaki K, Fujita H, Sato T, Kato S, Takahashi Y, Takeshita Y, Kanda T, Saito T, Saido TC, Hattori S, Hozumi Y, Yamada Y, Waki H. GLP-1 receptor signaling restores aquaporin 4 subcellular polarization in reactive astrocytes and promotes amyloid β clearance in a mouse model of Alzheimer's disease. Biochem Biophys Res Commun 2024; 741:151016. [PMID: 39577079 DOI: 10.1016/j.bbrc.2024.151016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 11/16/2024] [Indexed: 11/24/2024]
Abstract
The physiological actions of a gut hormone, glucagon-like peptide-1 (GLP-1), in Alzheimer's disease (AD) brain remain poorly understood, although GLP-1 receptor (GLP-1R) expression in this organ has been shown in several experimental studies. Therefore, we explored whether the GLP-1R signaling promotes the clearance of amyloid β (Aβ) (1-42) which is a core pathological hallmark of AD, focusing on the water channel protein aquaporin 4 (AQP4) localized to astrocyte endfeet perivascular membranes in intact brain. First, we confirmed that Glp1r mRNA is predominantly expressed at perivascular site of astrocytes in normal mouse cerebral cortex through in situ hybridization analysis. Next, we observed that 20-week subcutaneous administration of a GLP-1R agonist (GLP-1RA) liraglutide significantly reduced Aβ (1-42) accumulation in the cerebral cortex and improved spatial working memory in an AD mouse model, AppNL-G-F/NL-G-F mice. Furthermore, our current data revealed that the 4-week liraglutide treatment relocalized subcellular AQP4 in morphologically injured reactive astrocytes of AppNL-G-F/NL-G-F mice to the cell surface perivascular site through PKA-mediated AQP4 phosphorylation. Such translocation of phosphorylated AQP4 to astrocyte cell surface following incubation with liraglutide was observed also in the present in vitro study using the cell line in which AQP4 cDNA was introduced into immortalized human astrocyte. These results suggest that enhanced intracerebral GLP-1R signaling following peripheral administration of GLP-1RA restores AQP4 subcellular polarization in reactive astrocytes and would promote Aβ excretion possibly through increasing AQP4-mediated intracerebral water flux in the brain in AD.
Collapse
Affiliation(s)
- Kana Sasaki
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan
| | - Hiroki Fujita
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan.
| | - Takehiro Sato
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shunske Kato
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan; Center for Medical Education and Training, Akita University Hospital, Akita, Japan
| | - Yuya Takahashi
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yukio Takeshita
- Blood-Brain Barrier Research Center, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan
| | - Takashi Kanda
- Department of Clinical Neuroscience, Yamaguchi University Graduate School of Medicine, Ube, Yamaguchi, Japan; Neuromuscular Center Yoshimizu Hospital, Shimonoseki, Yamaguchi, Japan
| | - Takashi Saito
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan; Department of Neurocognitive Science, Institute of Brain Science, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi, Japan
| | - Takamori C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, Wako, Saitama, Japan
| | - Satoko Hattori
- Division of Systems Medical Science, Center for Medical Science, Fujita Health University, Toyoake, Aichi, Japan; Research Creation Support Center, Aichi Medical University, Nagakute, Aichi, Japan
| | - Yasukazu Hozumi
- Department of Cell Biology and Morphology, Akita University Graduate School of Medicine, Akita, Japan
| | - Yuichiro Yamada
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan; Center for Diabetes, Endocrinology and Metabolism, Kansai Electric Power Hospital, Osaka, Japan; Yutaka Seino Distinguished Center for Diabetes Research, Kansai Electric Power Medical Research Institute, Kyoto, Japan.
| | - Hironori Waki
- Department of Metabolism and Endocrinology, Akita University Graduate School of Medicine, Akita, Japan.
| |
Collapse
|
21
|
Sugahara C, Kin K, Sasaki T, Sasada S, Kawauchi S, Yabuno S, Nagase T, Hirayama T, Masai K, Hosomoto K, Okazaki Y, Kawai K, Tanimoto S, Hirata Y, Miyake H, Naito H, Yasuhara T, Borlongan CV, Date I, Tanaka S. Repeated non-hemorrhagic and non-contusional mild traumatic brain injury in rats elicits behavioral impairment with microglial activation, astrogliosis, and tauopathy: Reproducible and quantitative model of chronic traumatic encephalopathy. Brain Res 2024:149412. [PMID: 39743034 DOI: 10.1016/j.brainres.2024.149412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 11/30/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Chronic traumatic encephalopathy (CTE) has attracted attention due to sports-related head trauma or repetitive mild traumatic brain injury (mTBI). However, the pathology of CTE remains underexplored. Reproducible and quantitative model of CTE has yet to be established. The aim of this study is to establish a highly reproducible model of CTE with behavioral and histological manifestations. First, the pathological symptoms of mTBI with no intracranial hemorrhage or contusion using the weight drop model of 52 g ball from a height of 30 cm was determined using hematoxylin and eosin staining. Adult rats that received single, double, or triple head impacts were compared with sham behaviorally and histologically. Results revealed that rats exposed to repetitive mTBI showed motor impairment with gradual recovery over time, which was prolonged as the number of head impact increased. Similarly, cognitive function was impaired by repetitive mTBI and the recovery depended on the number of head impact. Histologically, GFAP positive astrocytes increased with repetitive mTBI, although Iba-1 positive microglial aggregation was limited. At 4w, phosphorylated Tau significantly accumulated in the prefrontal cortex, corpus callosum, CA1, and dentate gyrus of rats that received triple mTBI, compared to sham or those exposed to single, or double mTBI. This repetitive mTBI rat model provides a highly reproducible and quantifiable brain and behavioral pathology reminiscent of CTE.
Collapse
Affiliation(s)
- Chiaki Sugahara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kyohei Kin
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Tatsuya Sasaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Susumu Sasada
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoshi Kawauchi
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Satoru Yabuno
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takayuki Nagase
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takahiro Hirayama
- Department of Emergency, Critical Care, and Disaster Medicine, Okayama University Graduate School of Medicine, Dentistry, and Pharmaceutical Sciences, Okayama, Japan
| | - Kaori Masai
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Kakeru Hosomoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yosuke Okazaki
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Koji Kawai
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shun Tanimoto
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Yuichi Hirata
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hayato Miyake
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiromichi Naito
- Department of Medical Neurobiology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Takao Yasuhara
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan.
| | - Cesar V Borlongan
- Department of Neurosurgery and Brain Repair, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Isao Date
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Shota Tanaka
- Department of Neurological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| |
Collapse
|
22
|
Sokolova D, Ghansah SA, Puletti F, Georgiades T, De Schepper S, Zheng Y, Crowley G, Wu L, Rueda-Carrasco J, Koutsiouroumpa A, Muckett P, Freeman OJ, Khakh BS, Hong S. Astrocyte-derived MFG-E8 facilitates microglial synapse elimination in Alzheimer's disease mouse models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.31.606944. [PMID: 39257734 PMCID: PMC11383703 DOI: 10.1101/2024.08.31.606944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Region-specific synapse loss is an early pathological hallmark in Alzheimer's disease (AD). Emerging data in mice and humans highlight microglia, the brain-resident macrophages, as cellular mediators of synapse loss; however, the upstream modulators of microglia-synapse engulfment remain elusive. Here, we report a distinct subset of astrocytes, which are glial cells essential for maintaining synapse homeostasis, appearing in a region-specific manner with age and amyloidosis at onset of synapse loss. These astrocytes are distinguished by their peri-synaptic processes which are 'bulbous' in morphology, contain accumulated p62-immunoreactive bodies, and have reduced territorial domains, resulting in a decrease of astrocyte-synapse coverage. Using integrated in vitro and in vivo approaches, we show that astrocytes upregulate and secrete phagocytic modulator, milk fat globule-EGF factor 8 (MFG-E8), which is sufficient and necessary for promoting microglia-synapse engulfment in their local milieu. Finally, we show that knocking down Mfge8 specifically from astrocytes using a viral CRISPR-saCas9 system prevents microglia-synapse engulfment and ameliorates synapse loss in two independent amyloidosis mouse models of AD. Altogether, our findings highlight astrocyte-microglia crosstalk in determining synapse fate in amyloid models and nominate astrocytic MFGE8 as a potential target to ameliorate synapse loss during the earliest stages of AD.
Collapse
Affiliation(s)
- Dimitra Sokolova
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
- Neuroscience BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Shari Addington Ghansah
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Francesca Puletti
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Tatiana Georgiades
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Sebastiaan De Schepper
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Yongjing Zheng
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Gerard Crowley
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Ling Wu
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Javier Rueda-Carrasco
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Angeliki Koutsiouroumpa
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Philip Muckett
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| | - Oliver J. Freeman
- Neuroscience BioPharmaceuticals R&D, AstraZeneca, Cambridge, United Kingdom
| | - Baljit S. Khakh
- Department of Physiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA; Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095-1751, USA
| | - Soyon Hong
- UK Dementia Research Institute, Institute of Neurology, University College London, Gower Street, London, WC1E 6BT, United Kingdom
| |
Collapse
|
23
|
Isasi E, Olivera-Bravo S. Neurovascular unit impairment in iron deficiency anemia. Neuroscience 2024:S0306-4522(24)00757-7. [PMID: 39733822 DOI: 10.1016/j.neuroscience.2024.12.050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/16/2024] [Accepted: 12/26/2024] [Indexed: 12/31/2024]
Abstract
Iron is one of the crucial elements for CNS development and function and its deficiency (ID) is the most common worldwide nutrient deficit in the world. Iron deficiency anemia (IDA) in pregnant women and infants is a worldwide health problem due to its high prevalence and its irreversible long-lasting effects on brain development. Even with iron supplementation, IDA during pregnancy and/or breastfeeding can result in irreversible cognitive, motor, and behavioral impairments. The neurovascular unit (NVU) plays an important role in iron transport within the CNS as well as in the blood brain-barrier (BBB) formation and maturation, vasculogenesis/angiogenesis, neurovascular coupling and metabolic waste clearance. In animal models of IDA, significant changes have been observed at the capillary level, including alterations in iron transport, vasculogenesis, astrocyte endfeet, and pericytes. Despite these findings, the role of the NVU in IDA remains poorly understood. This review summarizes the potential effects of ID/IDA on brain development, myelination and neuronal function and discusses the role of NVU cells in iron metabolism, BBB, vasculogenesis/angiogenesis, neurovascular coupling and metabolic waste clearance. Furthermore, it emphasizes the need to view the NVU as a whole and as a potential target for ID/IDA. However, it remains unclear to what extent NVU alterations contribute to neuronal dysfunction, myelination abnormalities, and synaptic disturbances described in IDA.
Collapse
Affiliation(s)
- Eugenia Isasi
- Unidad Académica de Histología y Embriología, Facultad de Medicina, UdelaR, Montevideo, Uruguay; Departamento de Neurobiología y Neuropatología, IIBCE, MEC, Montevideo, Uruguay
| | - Silvia Olivera-Bravo
- Departamento de Neurobiología y Neuropatología, IIBCE, MEC, Montevideo, Uruguay.
| |
Collapse
|
24
|
Li S, Chen Y, Chen G. Cognitive disorders: potential astrocyte-based mechanism. Brain Res Bull 2024:111181. [PMID: 39725239 DOI: 10.1016/j.brainresbull.2024.111181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/28/2024]
Abstract
Cognitive disorders are a common clinical manifestation, including a deterioration in the patient's memory ability, attention, executive power, language, and other functions. The contributing factors of cognitive disorders are numerous and diverse in nature, including organic diseases and other mental disorders. Neurodegenerative diseases are a common type of organic disease related to the pathology of neuronal death and disruption of glial cell balance, ultimately accompanied with cognitive impairment. Thus, cognitive disorder frequently serves as an extremely critical indicator of neurodegenerative disorders. Cognitive impairments negatively affect patients' daily lives. However, our understanding of the precise pathogenic pathways of cognitive defects remains incomplete. The most prevalent kind of glial cells in the central nervous system are called astrocytes. They have a unique significance in cerebral function because of their wide range of functions in maintaining homeostasis in the central nervous system, regulating synaptic plasticity, and so on. Dysfunction of astrocytes is intimately linked to cognitive disorders, and we are attempting to understand this phenomenon predominantly from those perspectives: neuroinflammation, astrocytic senescence, connexin, Ca2+ signaling, mitochondrial dysfunction, and the glymphatic system.
Collapse
Affiliation(s)
- Shiyu Li
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Yeru Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Gang Chen
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| |
Collapse
|
25
|
Marques SI, Sá SI, Carmo H, Carvalho F, Silva JP. Pharmaceutical-mediated neuroimmune modulation in psychiatric/psychological adverse events. Prog Neuropsychopharmacol Biol Psychiatry 2024; 135:111114. [PMID: 39111563 DOI: 10.1016/j.pnpbp.2024.111114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/21/2024] [Accepted: 08/03/2024] [Indexed: 08/13/2024]
Abstract
The therapeutic use of many pharmaceuticals, including small molecules and biological therapies, has been associated with the onset of psychiatric and psychological adverse events (PPAEs), posing substantial concerns to patients' health and safety. These events, which encompass mood (e.g., depression, schizophrenia, suicidal ideation) and cognitive changes (e.g., learning and memory impairment, dementia) often remain undetected until advanced stages of clinical trials or pharmacovigilance, mostly because the mechanisms underlying the onset of PPAEs remain poorly understood. In recent years, the role of neuroimmune modulation (comprising an intricate interplay between various cell types and signaling pathways) in PPAEs has garnered substantial interest. Indeed, understanding these complex interactions would substantially contribute to increase the ability to predict the potential onset of PPAEs during preclinical stages of a new drug's R&D. This review provides a comprehensive summary of the most recent advances in neuroimmune modulation-related mechanisms contributing to the onset of PPAEs and their association with specific pharmaceuticals. Reported data strongly support an association between neuroimmune modulation and the onset of PPAEs. Pharmaceuticals may target specific molecular pathways and pathway elements (e.g., cholinergic and serotonergic systems), which in turn may directly or indirectly impact the inflammatory status and the homeostasis of the brain, regulating inflammation and neuronal function. Also, modulation of the peripheral immune system by pharmaceuticals that do not permeate the blood-brain barrier (e.g., monoclonal antibodies) may alter the neuroimmunomodulatory status of the brain, leading to PPAEs. In summary, this review underscores the diverse pathways through which drugs can influence brain inflammation, shedding light on potential targeted interventions.
Collapse
Affiliation(s)
- Sandra I Marques
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Susana I Sá
- Unit of Anatomy, Department of Biomedicine, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal; CINTESIS@RISE, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal.
| | - Helena Carmo
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - Félix Carvalho
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| | - João P Silva
- UCIBIO - Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal; i4HB - Institute for Health and Bioeconomy, Faculty of Pharmacy, University of Porto, 4050-313 Porto, Portugal.
| |
Collapse
|
26
|
Kurogi Y, Sanagi T, Ono D, Tsunematsu T. Chemogenetic activation of astrocytes modulates sleep-wakefulness states in a brain region-dependent manner. SLEEP ADVANCES : A JOURNAL OF THE SLEEP RESEARCH SOCIETY 2024; 5:zpae091. [PMID: 39717113 PMCID: PMC11664484 DOI: 10.1093/sleepadvances/zpae091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 12/04/2024] [Indexed: 12/25/2024]
Abstract
Study Objectives Astrocytes change their intracellular calcium (Ca2+) concentration during sleep/wakefulness states in mice. Furthermore, the Ca2+ dynamics in astrocytes vary depending on the brain region. However, it remains unclear whether alterations in astrocyte activity can affect sleep-wake states and cortical oscillations in a brain region-dependent manner. Methods Astrocyte activity was artificially manipulated in mice using chemogenetics. Astrocytes in the hippocampus and pons, which are 2 brain regions previously classified into different clusters based on their Ca2+ dynamics during sleep-wakefulness, were focused on to compare whether there are differences in the effects of astrocytes from different brain regions. Results The chemogenetic activation of astrocytes in the hippocampus significantly decreased the total time of wakefulness and increased the total time of sleep. This had little effect on cortical oscillations in all sleep-wakefulness states. On the other hand, the activation of astrocytes in the pons substantially suppressed rapid eye movement (REM) sleep in association with a decreased number of REM episodes, indicating strong inhibition of REM onset. Regarding cortical oscillations, the delta wave component during non-REM sleep was significantly enhanced. Conclusions These results suggest that astrocytes modulate sleep-wakefulness states and cortical oscillations. Furthermore, the role of astrocytes in sleep-wakefulness states appears to vary among brain regions.
Collapse
Affiliation(s)
- Yuta Kurogi
- Graduate School of Life Sciences, Tohoku University, Sendai, Japan
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
| | - Tomomi Sanagi
- Creative Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| | - Daisuke Ono
- Stress Recognition and Response, Research Institute of Environmental Medicine, Nagoya University, Nagoya, Japan
- Department of Neural Regulation, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tomomi Tsunematsu
- Department of Biological Sciences, Faculty of Science, Hokkaido University, Sapporo, Japan
- Creative Interdisciplinary Research Division, Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Japan
| |
Collapse
|
27
|
Qiu Y, Lu G, Zhang S, Minping L, Xue X, Junyu W, Zheng Z, Qi W, Guo J, Zhou D, Huang H, Deng Z. Mitochondrial dysfunction of Astrocyte induces cell activation under high salt condition. Heliyon 2024; 10:e40621. [PMID: 39660210 PMCID: PMC11629238 DOI: 10.1016/j.heliyon.2024.e40621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/20/2024] [Accepted: 11/20/2024] [Indexed: 12/12/2024] Open
Abstract
Excess dietary sodium can accumulate in brain and adversely affect human health. We have confirmed in previous studies that high salt can induce activation of astrocyte manifested by the secretion of various inflammatory factors. In order to further explore the effect of high salt on the internal cell metabolism of astrocytes, RNA sequencing was performed on astrocytes under high salt environment, which indicated the oxidative phosphorylation and glycolysis pathways of astrocytes were downregulated. Next, we found that high salt concentrations elicited astrocyte mitochondrial morphology change, as evidenced by swelling from a short rod to a round shape through a High Intelligent and Sensitive Structured Illumination Microscope (HIS-SIM). Furthermore, we found that high salt concentrations reduced astrocyte mitochondrial oxygen consumption and membrane potential. Treatment with 18-kDa translocator protein (TSPO) ligands FGIN-1-27 improved mitochondrial networks and reversed astrocyte activation under high-salt circumstances. Our study shows that high salt can directly disrupt astrocytic mitochondrial homeostasis and function. Targeting translocator protein signaling may have therapeutic potential against high-salt neurotoxicity.
Collapse
Affiliation(s)
- Yuemin Qiu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
- Department of Neurology, Shenzhen Bao'an District Songgang People's Hospital, No.2 Shajiang Road, Shenzhen, 518100, China
| | - Gengxin Lu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Shifeng Zhang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Li Minping
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Xu Xue
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Wu Junyu
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zhihui Zheng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Weiwei Qi
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Junjie Guo
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Dongxiao Zhou
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Haiwei Huang
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| | - Zhezhi Deng
- Department of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangdong Provincial Key Laboratory of Diagnosis and Treatment of Major Neurological Diseases, National Key Clinical Department and Key Discipline of Neurology, No.58 Zhongshan Road 2, Guangzhou, 510080, China
| |
Collapse
|
28
|
Huang Z, Xu P, Hess DC, Zhang Q. Cellular senescence as a key contributor to secondary neurodegeneration in traumatic brain injury and stroke. Transl Neurodegener 2024; 13:61. [PMID: 39668354 PMCID: PMC11636056 DOI: 10.1186/s40035-024-00457-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Traumatic brain injury (TBI) and stroke pose major health challenges, impacting millions of individuals globally. Once considered solely acute events, these neurological conditions are now recognized as enduring pathological processes with long-term consequences, including an increased susceptibility to neurodegeneration. However, effective strategies to counteract their devastating consequences are still lacking. Cellular senescence, marked by irreversible cell-cycle arrest, is emerging as a crucial factor in various neurodegenerative diseases. Recent research further reveals that cellular senescence may be a potential driver for secondary neurodegeneration following brain injury. Herein, we synthesize emerging evidence that TBI and stroke drive the accumulation of senescent cells in the brain. The rationale for targeting senescent cells as a therapeutic approach to combat neurodegeneration following TBI/stroke is outlined. From a translational perspective, we emphasize current knowledge and future directions of senolytic therapy for these neurological conditions.
Collapse
Affiliation(s)
- Zhihai Huang
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA, 71103, USA
| | - Peisheng Xu
- Department of Drug Discovery and Biomedical Sciences, College of Pharmacy, University of South Carolina, 715 Sumter, Columbia, SC, 29208, USA
| | - David C Hess
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA
| | - Quanguang Zhang
- Department of Neurology, Medical College of Georgia, Augusta University, 1120 15th Street, Augusta, GA, 30912, USA.
| |
Collapse
|
29
|
Chauhan P, Begum MY, Narapureddy BR, Gupta S, Wadhwa K, Singh G, Kumawat R, Sharma N, Ballal S, Jha SK, Abomughaid MM, B D, Ojha S, Jha NK. Unveiling the Involvement of Herpes Simplex Virus-1 in Alzheimer's Disease: Possible Mechanisms and Therapeutic Implications. Mol Neurobiol 2024:10.1007/s12035-024-04535-4. [PMID: 39648189 DOI: 10.1007/s12035-024-04535-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 10/01/2024] [Indexed: 12/10/2024]
Abstract
Viruses pose a significant challenge and threat to human health, as demonstrated by the current COVID-19 pandemic. Neurodegeneration, particularly in the case of Alzheimer's disease (AD), is significantly influenced by viral infections. AD is a neurodegenerative disease that affects people of all ages and poses a significant threat to millions of individuals worldwide. The precise mechanism behind its development is not yet fully understood; however, the emergence and advancement of AD can be hastened by various environmental factors, such as bacterial and viral infections. There has been a longstanding suspicion that the herpes simplex virus-1 (HSV-1) may have a role to play in the development or advancement of AD. Reactivation of HSV-1 could potentially lead to damage to neurons, either by direct means or indirectly by triggering inflammation. This article provides an overview of the connection between HSV-1 infections and immune cells (astrocytes, microglia, and oligodendrocytes) in the progression of AD. It summarizes recent scientific research on how HSV-1 affects neurons, which could potentially shed light on the clinical features and treatment options for AD. In addition, the paper has explored the impact of HSV-1 on neurons and its role in various aspects of AD, such as Aβ secretion, tau hyperphosphorylation, metabolic dysregulation, oxidative damage, apoptosis, and autophagy. It is believed that the immune response triggered by HSV-1 reactivation plays a role in the development of neurodegeneration in AD. Despite the lack of a cure for AD, researchers have made significant efforts to study the clinical and pathological aspects of the disease, identify biomarkers, and gain insight into its underlying causes. The goal is to achieve early diagnosis and develop treatments that can modify the progression of the disease. The current article discusses the most promising therapy for combating the viral impacts, which provides additional evidence for the frequent reactivations of latent HSV-1 in the AD brain. However, further research is still required to establish the molecular and cellular mechanisms underlying the development of AD through the reactivation of HSV-1. This could potentially lead to new insights in drug development aimed at preventing HSV-1 reactivation and the subsequent development and progression of AD.
Collapse
Affiliation(s)
- Payal Chauhan
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - M Yasmin Begum
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha, Saudi Arabia
| | - Bayapa Reddy Narapureddy
- Department of Public Health, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Karan Wadhwa
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India
| | - Govind Singh
- Department of Pharmaceutical Sciences, Maharshi Dayanand University, Rohtak, 124001, India.
| | - Rohit Kumawat
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajsthan, Jaipur, India
| | - Naveen Sharma
- Chandigarh Pharmacy College, Chandigarh Group of Colleges Jhanjeri, Mohali, 140307, Punjab, India
| | - Suhas Ballal
- Departmant of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Saurabh Kumar Jha
- Department of Zoology, Kalindi College, University of Delhi, Delhi, 110008, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, 61922, Bisha, Saudi Arabia
| | - Dheepak B
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Shreesh Ojha
- Department of Pharmacology and Therapeutics, College of Medicine and Health Sciences, United Arab Emirates University, P.O. Box 15551, Al Ain, United Arab Emirates
| | - Niraj Kumar Jha
- Department of Biotechnology, School of Biosciences & Technology, Galgotias University, Greater Noida, India.
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India.
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, 144411, India.
| |
Collapse
|
30
|
Wang M, Li T, Li W, Song T, Zhao C, Wu Q, Cui W, Hao Y, Hou Y, Zhu P. Unraveling the neuroprotective potential of scalp electroacupuncture in ischemic stroke: A key role for electrical stimulation. Neuroscience 2024; 562:160-181. [PMID: 39401739 DOI: 10.1016/j.neuroscience.2024.10.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/23/2024] [Accepted: 10/09/2024] [Indexed: 11/05/2024]
Abstract
This study aims to explore the neuroprotective effects of scalp Electroacupuncture (EA) on ischemic stroke, with a specific focus on the role of electrical stimulation (ES). Employing a rat model of middle cerebral artery occlusion (MCAO), we used methods such as Triphenyl tetrazolium chloride staining, micro-CT scanning, Enzyme linked immunosorbent assay (ELISA), and immunofluorescence to assess the impacts of EA. We further conducted RNA-seq analysis and in vitro experiments with organotypic brain slices and cerebral organoids to explore the underlying mechanisms. Our research revealed that EA notably reduced cerebral infarct volume and improved regional cerebral blood flow in rats following MCAO. Micro-CT imaging showed improved vascular integrity in EA-treated groups. Histological analyses, including HE staining, indicated reduced brain tissue damage. ELISA demonstrated a decrease in pro-inflammatory cytokines TNF-α, IL-1β, and IL-6, suggesting improved blood-brain barrier function. Immunofluorescence and Western blot analyses revealed that EA treatment significantly inhibited microglial and astrocytic overactivation. RNA-seq analysis of brain tissues highlighted a downregulation of immune pathways and inflammatory responses, confirming the neuroprotective role of EA. This was further corroborated by in vitro experiments using organotypic brain slices and cerebral organoids, which showcased the efficacy of electrical stimulation in reducing neuroinflammation and protecting neuronal cells. The study highlights the potential of scalp EA, particularly its ES component, in treating ischemic stroke. It provides new insights into the mechanisms of EA, emphasizing its efficacy in neuroprotection and modulation of neuroinflammation, and suggests avenues for optimized treatment strategies in stroke therapy.
Collapse
Affiliation(s)
- Mingye Wang
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Tongtong Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Wenyan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China
| | - Tao Song
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Chi Zhao
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Qiulan Wu
- Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China
| | - Wenwen Cui
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yuanyuan Hao
- Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China
| | - Yunlong Hou
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, No.326, Xinshi South Road, Shijiazhuang 050091, Hebei, China; Shijiazhuang Yiling Pharmaceutical Co., Ltd, New Drug Evaluation Center, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China; Hebei Medical University, No.361 Zhongshan East Road, Shijiazhuang 050011, Hebei, China; National Key Laboratory for Innovation and Transformation of Luobing Theory, No.238, the South of Tianshan Street, Shijiazhuang 050035, Hebei, China; Key Laboratory of State Administration of TCM Cardio-Cerebral Vessel Collateral Disease, No.238, the South of Tianshan Street, Shijiazhuang, 050035, Hebei, China.
| | - Pengyu Zhu
- The Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 411, The Street of Guogeli, Harbin 150001, Heilongjiang, China.
| |
Collapse
|
31
|
Gong X, Cai W, Yang D, Wang W, Che H, Li H. Effect of the arabinogalactan from Ixeris chinensis (Thunb.) Nakai. attenuates DSS-induced colitis and accompanying depression-like behavior. Int J Biol Macromol 2024; 286:138525. [PMID: 39647733 DOI: 10.1016/j.ijbiomac.2024.138525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/21/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
An arabinogalactan (ICPA) was extracted from the medicinal and edible plant Ixeris chinensis (Thunb.) Nakai., and ICPA exhibited excellent immunomodulatory activity. In this research, the impact of ICPA on DSS-induced ulcerative colitis was investigated. The results indicated that ICPA ameliorated the symptoms of colitis mice including loss of body weight, decrease of disease activity index, shortness of colon length and reduction of spleen index that caused by DSS. After treatment with ICPA, inflammatory cell infiltration and crypt loss were alleviated, and the number of goblet epithelial cells was enriched. ICPA inhibited the overproduction of TNF-α, IL-1β, and NLRP3, and promoted the secretion of IL-10 in colon tissues. Meanwhile, the intestinal barrier integrity was restored through increasing the expression of ZO-1 and occludin. ICPA could also regulate the structure of gut microbiota through elevating the abundance of Turicibacter and Bifidobacterium, and decreasing the ratio of Bacteroidetes/Firmicutes. In addition, ICPA improved the depression-like behavior of UC mice, and reduced the expression of proteins NLRP3, GFAP, and Iba-1 in brain tissues. These results suggested ICPA had an alleviative effect on UC and accompanied depression-like behavior, and could be developed as a dietary supplement for the prevention and treatment of UC.
Collapse
Affiliation(s)
- Xinwei Gong
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Wanshuang Cai
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Dezhao Yang
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Wei Wang
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Hongxia Che
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China
| | - Hongyan Li
- College of Marine Science and Biological Engineering, Shandong Provincial Key Laboratory of Biochemical Engineering, Qingdao University of Science and Technology, Qingdao 266042, China.
| |
Collapse
|
32
|
Tian L, Chen J, Liu X, Wei Y, Zhao Y, Shi Y, Li K, Liu H, Lai W, Lin B. Prenatal exposure on nanoplastics: A study of spatial transcriptomics in hippocampal offspring. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 366:125480. [PMID: 39644950 DOI: 10.1016/j.envpol.2024.125480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/17/2024] [Accepted: 12/04/2024] [Indexed: 12/09/2024]
Abstract
Nanoplastics, as environmental contaminants, are thought to have irreversible impacts on the developing brains of infants and early children; however, the degree of the effects and the mechanisms of damage are unknown. In this study, spatial transcriptomics was used to investigate changes in the hippocampal region of rats descended from maternal exposure to polystyrene nanoplastics (PS-NPs), and the transcriptomes of each spot were sequenced, allowing us to visualize the hippocampus's transcriptional landscape as well as clarify the gene expression profiles of each cell type. Spatial transcriptomics was used to explore changes in the hippocampus region of rats exposed to PS-NPs during brain formation and maturation.The study's findings showed that the offspring hippocampal region had fewer neurons, more astrocytes, and more excitatory neurons 1(ExN1). The pseudo-time study of astrocytes revealed a decrease in C3-type astrocytes and an increase in C2-type astrocytes. This finding raises the possibility that memory impairment in the offspring may result from the developmental obstruction of astrocytes following the intervention of PS-NPs. Moreover, the annotations of four hippocampus regions, CA1, CA2-3, DG, and HILUS, as well as the GO and GSVA of several cell types, revealed deficiencies that can contribute to learning memory impairment. The analysis suggested that decreased neuroglutamate (Glutamate) and γ-aminobutyric acid (GABA) secretion in offspring after PS-NPs intervention was associated with depression. Lastly, intercellular communication revealed alterations in several ligand receptor pathways associated with an increase in astrocytes. In conclusion, spatial transcriptomics reveals that maternal exposure to nanoplastics influences the development of the offspring's hippocampal brain and causes neurotoxicity, which accounts for the offspring's reduction in learning memory function.
Collapse
Affiliation(s)
- Lei Tian
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Jiang Chen
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China; School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Xuan Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Yizhe Wei
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Yiming Zhao
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China; School of Public Health, North China University of Science and Technology, Tangshan, 063200, China
| | - Yue Shi
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Kang Li
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Huanliang Liu
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Wenqing Lai
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China
| | - Bencheng Lin
- Military Medical Sciences Academy, Academy of Military Sciences, Tianjin Key Laboratory of Risk Assessment and Control Technology for Environment and Food Safety, Tianjin, 300050, China.
| |
Collapse
|
33
|
Cross K, Vetter SW, Alam Y, Hasan MZ, Nath AD, Leclerc E. Role of the Receptor for Advanced Glycation End Products (RAGE) and Its Ligands in Inflammatory Responses. Biomolecules 2024; 14:1550. [PMCID: PMC11673996 DOI: 10.3390/biom14121550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 01/03/2025] Open
Abstract
Since its discovery in 1992, the receptor for advanced glycation end products (RAGE) has emerged as a key receptor in many pathological conditions, especially in inflammatory conditions. RAGE is expressed by most, if not all, immune cells and can be activated by many ligands. One characteristic of RAGE is that its ligands are structurally very diverse and belong to different classes of molecules, making RAGE a promiscuous receptor. Many of RAGE ligands are damaged associated molecular patterns (DAMPs) that are released by cells under inflammatory conditions. Although RAGE has been at the center of a lot of research in the past three decades, a clear understanding of the mechanisms of RAGE activation by its ligands is still missing. In this review, we summarize the current knowledge of the role of RAGE and its ligands in inflammation.
Collapse
Affiliation(s)
| | | | | | | | | | - Estelle Leclerc
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, ND 58105, USA; (K.C.); (S.W.V.); (Y.A.); (M.Z.H.); (A.D.N.)
| |
Collapse
|
34
|
Villa J, Cury J, Kessler L, Tan X, Richter CP. Enhancing biocompatibility of the brain-machine interface: A review. Bioact Mater 2024; 42:531-549. [PMID: 39308547 PMCID: PMC11416625 DOI: 10.1016/j.bioactmat.2024.08.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/05/2024] [Accepted: 08/27/2024] [Indexed: 09/25/2024] Open
Abstract
In vivo implantation of microelectrodes opens the door to studying neural circuits and restoring damaged neural pathways through direct electrical stimulation and recording. Although some neuroprostheses have achieved clinical success, electrode material properties, inflammatory response, and glial scar formation at the electrode-tissue interfaces affect performance and sustainability. Those challenges can be addressed by improving some of the materials' mechanical, physical, chemical, and electrical properties. This paper reviews materials and designs of current microelectrodes and discusses perspectives to advance neuroprosthetics performance.
Collapse
Affiliation(s)
- Jordan Villa
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Joaquin Cury
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Lexie Kessler
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
| | - Xiaodong Tan
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
| | - Claus-Peter Richter
- Northwestern University-Feinberg School of Medicine, Department of Otolaryngology, USA
- The Hugh Knowles Center, Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Communication Sciences and Disorders, Northwestern University, USA
- Department of Biomedical Engineering, Northwestern University, USA
| |
Collapse
|
35
|
Sámano C, Mazzone GL. The role of astrocytes response triggered by hyperglycaemia during spinal cord injury. Arch Physiol Biochem 2024; 130:724-741. [PMID: 37798949 DOI: 10.1080/13813455.2023.2264538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 09/22/2023] [Indexed: 10/07/2023]
Abstract
OBJECTIVE This manuscript aimed to provide a comprehensive overview of the physiological, molecular, and cellular mechanisms triggered by reactive astrocytes (RA) in the context of spinal cord injury (SCI), with a particular focus on cases involving hyperglycaemia. METHODS The compilation of articles related to astrocyte responses in neuropathological conditions, with a specific emphasis on those related to SCI and hyperglycaemia, was conducted by searching through databases including Science Direct, Web of Science, and PubMed. RESULTS AND CONCLUSIONS This article explores the dual role of astrocytes in both neurophysiological and neurodegenerative conditions within the central nervous system (CNS). In the aftermath of SCI and hyperglycaemia, astrocytes undergo a transformation into RA, adopting a distinct phenotype. While there are currently no approved therapies for SCI, various therapeutic strategies have been proposed to alleviate the detrimental effects of RAs following SCI and hyperglycemia. These strategies show promising potential in the treatment of SCI and its likely comorbidities.
Collapse
Affiliation(s)
- C Sámano
- Departamento de Ciencias Naturales, Universidad Autónoma Metropolitana, Unidad Cuajimalpa (UAM-C), Ciudad de México, México
| | - G L Mazzone
- Instituto de Investigaciones en Medicina Traslacional (IIMT), CONICET-Universidad Austral, Pilar, Buenos Aires, Argentina
- Facultad de Ciencias Biomédicas, Universidad Austral, Pilar, Buenos Aires, Argentina
| |
Collapse
|
36
|
Chen W, Wu Z, Yin M, Zhang Y, Qin Y, Liu X, Tu J. Blockage of p38MAPK in astrocytes alleviates brain damage in a mouse model of embolic stroke through the CX43/AQP4 axis. J Stroke Cerebrovasc Dis 2024; 33:108085. [PMID: 39393507 DOI: 10.1016/j.jstrokecerebrovasdis.2024.108085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND Cerebral edema, a significant complication arising from acute ischemic stroke (IS), has a critical influence on morbidity and mortality. p38MAPK has been shown to promote neuronal apoptosis and brain damage. However, the role of the p38MAPK inhibitor SKF-86002 in protecting against ischemic injury and cerebral edema remains unclear. METHODS Infarct area was examined by TTC staining in middle cerebral artery occlusion (MCAO) mice. Neurological score and brain water content were evaluated. TUNEL and NeuN staining were used to assess neuronal apoptosis and the survival of neurons. Blood-brain barrier (BBB) permeability was determined by Evans blue. Double immunofluorescence staining detected the colocalization of AQP4 and CX43 in astrocytes. IHC staining revealed CX43 and AQP4 expression. EDU staining detected the proliferation of Oxygen and glucose deprivation/reoxygenation (OGD/R)-treated astrocytes. Levels of oxidative stress markers were determined using commercial kits. ELISA was used to assess the secretion of pro-inflammatory factors. RT-qPCR measured the expression of CX43, AQP4 and pro-inflammatory factors. Western blot analyzed the levels of p-p38/p38, AQP4 and CX43. Co-immunoprecipitation (Co-IP) determined the interaction between CX43 and AQP4. RESULTS SKF-86002 attenuated brain damage, edema, and neuronal apoptosis in MCAO mice. Astrocyte proliferation was suppressed, and oxidative stress and inflammation were alleviated by SKF-86002 treatment. SKF-86002 negatively regulated p38 signaling and the expression of AQP4 and CX43. Additionally, the expression of CX43/AQP4 within astrocytes was modulated by SKF-86002. CONCLUSION In summary, SKF-86002 alleviated IS injury and cerebral edema by inhibiting astrocyte proliferation, oxidative stress and inflammation. This effect was associated with the suppression of CX43/AQP4, suggesting that SKF-86002 shows promise as a novel therapeutic approach for preventing IS.
Collapse
Affiliation(s)
- Weiping Chen
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Institute of Neuroscience, Nanchang University, Nanchang 330031, Jiangxi Province, PR China; Jiangxi Provincial Clinical Medical Research Center for Neurological Disorders, Nanchang 330031, Jiangxi Province, PR China
| | - Zhiping Wu
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Institute of Neuroscience, Nanchang University, Nanchang 330031, Jiangxi Province, PR China; Jiangxi Provincial Clinical Medical Research Center for Neurological Disorders, Nanchang 330031, Jiangxi Province, PR China
| | - Min Yin
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Institute of Neuroscience, Nanchang University, Nanchang 330031, Jiangxi Province, PR China; Jiangxi Provincial Clinical Medical Research Center for Neurological Disorders, Nanchang 330031, Jiangxi Province, PR China
| | - Yangbo Zhang
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Institute of Neuroscience, Nanchang University, Nanchang 330031, Jiangxi Province, PR China; Jiangxi Provincial Clinical Medical Research Center for Neurological Disorders, Nanchang 330031, Jiangxi Province, PR China
| | - Yiren Qin
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, PR China
| | - Xu Liu
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Institute of Neuroscience, Nanchang University, Nanchang 330031, Jiangxi Province, PR China; Jiangxi Provincial Clinical Medical Research Center for Neurological Disorders, Nanchang 330031, Jiangxi Province, PR China.
| | - Jianglong Tu
- Department of Neurology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang 330006, Jiangxi Province, PR China; Institute of Neuroscience, Nanchang University, Nanchang 330031, Jiangxi Province, PR China; Jiangxi Provincial Clinical Medical Research Center for Neurological Disorders, Nanchang 330031, Jiangxi Province, PR China.
| |
Collapse
|
37
|
Barbosa GADC, Rubinho MP, Aquino-Júnior MK, Pedro JR, Donato LF, Trisciuzzi L, Silva AO, Ruginsk SG, Ceron CS, Peixoto N, Dias MVS, Pereira MGAG. Neuritogenesis and protective effects activated by Angiotensin 1-7 in astrocytes-neuron interaction. Neuropeptides 2024; 108:102480. [PMID: 39500142 DOI: 10.1016/j.npep.2024.102480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/17/2024] [Accepted: 10/18/2024] [Indexed: 11/18/2024]
Abstract
The renin angiotensin system (RAS) has been studied for its effects on various neurological disorders. The identification of functional receptors for Ang-(1-7) and Ang II peptides in astrocytes highlights the physiological modulation and the important role of these cells in the central nervous system. The present study aims to understand the role of RAS peptides, particularly Ang-(1-7) and Ang II, in the secretion of trophic factors by astrocytes and their effects on hippocampal neurons. We used primary cultures of astrocytes and neurons from the hippocampus of either sex neonate of Wistar strain rats. In the present study, we demonstrated that the treatment of astrocytes with Ang-(1-7) acts on the modulation of these cells, inducing reactive astrogliosis, identified through the increase in the expression of GFAP. Furthermore, we obtained a conditioned medium from astrocytes treated with Ang-(1-7), which in addition to promoting the secretion of neurotrophic factors essential for neuronal-glial interactions that are fundamental for neuritogenesis and neuronal survival, showed a neuroprotective effect against glutamatergic excitotoxicity. In turn, Ang II does not exhibit the same effects on astrocyte modulation, exacerbating deleterious effects on brain RAS. Neuron-astrocyte interactions have been shown to be an integral part of the central effects mediated by RAS, and this study has significantly contributed to the understanding of the biochemical mechanisms involved in the functioning of this system.
Collapse
Affiliation(s)
| | - Marina Prado Rubinho
- Department of Biochemistry, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | | | - Lívia Fligioli Donato
- Department of Biochemistry, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Leonardo Trisciuzzi
- Department of Biochemistry, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | | | - Silvia Graciela Ruginsk
- Department of Physiological Sciences, Biomedical Sciences Institute, Federal University of Alfenas, Alfenas, Minas Gerais, Brazil
| | - Carla Speroni Ceron
- Department of Biological Sciences, Federal University of Ouro Preto, Ouro Preto, Minas Gerais, Brazil
| | - Nathalia Peixoto
- Electrical & Computer Engineering Department, George Mason University, Fairfax, VA, United States of America
| | | | | |
Collapse
|
38
|
Hernández-Martín N, Martínez MG, Bascuñana P, Fernández de la Rosa R, García-García L, Gómez F, Solas M, Martín ED, Pozo MA. Astrocytic Ca 2+ activation by chemogenetics mitigates the effect of kainic acid-induced excitotoxicity on the hippocampus. Glia 2024; 72:2217-2230. [PMID: 39188024 DOI: 10.1002/glia.24607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 08/01/2024] [Accepted: 08/05/2024] [Indexed: 08/28/2024]
Abstract
Astrocytes play a multifaceted role regulating brain glucose metabolism, ion homeostasis, neurotransmitters clearance, and water dynamics being essential in supporting synaptic function. Under different pathological conditions such as brain stroke, epilepsy, and neurodegenerative disorders, excitotoxicity plays a crucial role, however, the contribution of astrocytic activity in protecting neurons from excitotoxicity-induced damage is yet to be fully understood. In this work, we evaluated the effect of astrocytic activation by Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) on brain glucose metabolism in wild-type (WT) mice, and we investigated the effects of sustained astrocyte activation following an insult induced by intrahippocampal (iHPC) kainic acid (KA) injection using 2-deoxy-2-[18F]-fluoro-D-glucose (18F-FDG) positron emission tomography (PET) imaging, along with behavioral test, nuclear magnetic resonance (NMR) spectroscopy and histochemistry. Astrocytic Ca2+ activation increased the 18F-FDG uptake, but this effect was not found when the study was performed in knock out mice for type-2 inositol 1,4,5-trisphosphate receptor (Ip3r2-/-) nor in floxed mice to abolish glucose transporter 1 (GLUT1) expression in hippocampal astrocytes (GLUT1ΔGFAP). Sustained astrocyte activation after KA injection reversed the brain glucose hypometabolism, restored hippocampal function, prevented neuronal death, and increased hippocampal GABA levels. The findings of our study indicate that astrocytic GLUT1 function is crucial for regulating brain glucose metabolism. Astrocytic Ca2+ activation has been shown to promote adaptive changes that significantly contribute to mitigating the effects of KA-induced damage. This evidence suggests a protective role of activated astrocytes against KA-induced excitotoxicity.
Collapse
Affiliation(s)
- Nira Hernández-Martín
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | | | - Pablo Bascuñana
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
| | - Rubén Fernández de la Rosa
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Bioimac, Universidad Complutense de Madrid, Madrid, Spain
| | - Luis García-García
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Francisca Gómez
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Departamento de Farmacología, Farmacognosia y Botánica, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid, Spain
| | - Maite Solas
- Facultad de Farmacia, Universidad de Navarra, Pamplona, Spain
| | | | - Miguel A Pozo
- Instituto Pluridisciplinar, Universidad Complutense de Madrid, Madrid, Spain
- Instituto de Investigación Sanitaria San Carlos (IdISSC), Hospital Clínico San Carlos, Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
39
|
Hannawi Y. Cerebral Small Vessel Disease: a Review of the Pathophysiological Mechanisms. Transl Stroke Res 2024; 15:1050-1069. [PMID: 37864643 DOI: 10.1007/s12975-023-01195-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
Cerebral small vessel disease (cSVD) refers to the age-dependent pathological processes involving the brain small vessels and leading to vascular cognitive impairment, intracerebral hemorrhage, and acute lacunar ischemic stroke. Despite the significant public health burden of cSVD, disease-specific therapeutics remain unavailable due to the incomplete understanding of the underlying pathophysiological mechanisms. Recent advances in neuroimaging acquisition and processing capabilities as well as findings from cSVD animal models have revealed critical roles of several age-dependent processes in cSVD pathogenesis including arterial stiffness, vascular oxidative stress, low-grade systemic inflammation, gut dysbiosis, and increased salt intake. These factors interact to cause a state of endothelial cell dysfunction impairing cerebral blood flow regulation and breaking the blood brain barrier. Neuroinflammation follows resulting in neuronal injury and cSVD clinical manifestations. Impairment of the cerebral waste clearance through the glymphatic system is another potential process that has been recently highlighted contributing to the cognitive decline. This review details these mechanisms and attempts to explain their complex interactions. In addition, the relevant knowledge gaps in cSVD mechanistic understanding are identified and a systematic approach to future translational and early phase clinical research is proposed in order to reveal new cSVD mechanisms and develop disease-specific therapeutics.
Collapse
Affiliation(s)
- Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University, 333 West 10th Ave, Graves Hall 3172C, Columbus, OH, 43210, USA.
| |
Collapse
|
40
|
Aghajani Mir M. Brain Fog: a Narrative Review of the Most Common Mysterious Cognitive Disorder in COVID-19. Mol Neurobiol 2024; 61:9915-9926. [PMID: 37874482 DOI: 10.1007/s12035-023-03715-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/14/2023] [Indexed: 10/25/2023]
Abstract
It has been more than three years since COVID-19 impacted the lives of millions of people, many of whom suffer from long-term effects known as long-haulers. Notwithstanding multiorgan complaints in long-haulers, signs and symptoms associated with cognitive characteristics commonly known as "brain fog" occur in COVID patients over 50, women, obesity, and asthma at excessive. Brain fog is a set of symptoms that include cognitive impairment, inability to concentrate and multitask, and short-term and long-term memory loss. Of course, brain fog contributes to high levels of anxiety and stress, necessitating an empathetic response to this group of COVID patients. Although the etiology of brain fog in COVID-19 is currently unknown, regarding the mechanisms of pathogenesis, the following hypotheses exist: activation of astrocytes and microglia to release pro-inflammatory cytokines, aggregation of tau protein, and COVID-19 entry in the brain can trigger an autoimmune reaction. There are currently no specific tests to detect brain fog or any specific cognitive rehabilitation methods. However, a healthy lifestyle can help reduce symptoms to some extent, and symptom-based clinical management is also well suited to minimize brain fog side effects in COVID-19 patients. Therefore, this review discusses mechanisms of SARS-CoV-2 pathogenesis that may contribute to brain fog, as well as some approaches to providing therapies that may help COVID-19 patients avoid annoying brain fog symptoms.
Collapse
Affiliation(s)
- Mahsa Aghajani Mir
- Deputy of Research and Technology, Babol University of Medical Sciences, Babol, Iran.
| |
Collapse
|
41
|
Shrivastava V, Tyagi S, Dey D, Singh A, Palanichamy JK, Sinha S, Sharma JB, Seth P, Sen S. Glial cholesterol redistribution in hypoxic injury in vitro influences oligodendrocyte maturation and myelination. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167476. [PMID: 39181517 DOI: 10.1016/j.bbadis.2024.167476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 08/16/2024] [Accepted: 08/16/2024] [Indexed: 08/27/2024]
Abstract
Hypoxic insult to the fetal brain causes loss of vulnerable premyelinating oligodendrocytes and arrested oligodendrocyte differentiation. Astrocytes influence oligodendrocyte differentiation and the astrocytic response to hypoxia could affect oligodendrocyte maturation under hypoxia. To identify pathways by which astrocytes influence oligodendroglial maturation in hypoxic injury, human fetal neural stem cell-derived astrocytes were exposed to 0.2 % oxygen for 48 hours. Transcriptomic analysis revealed the upregulation of the cholesterol-biosynthesis pathway in hypoxia-exposed astrocytes. Hypoxia-exposed primary astrocytes and astrocytic cell line (SVG) showed increased expression of hydroxy-methyl-glutaryl-CoA reductase (HMGCR), squalene epoxidase (SQLE), apolipoprotein E (apoE) and ATP-binding cassette transporter 1 (ABCA1) on qPCR and Western blot. Hypoxic SVG also showed increased cholesterol content in cells and culture supernatants and increased cell surface expression of ABCA1. Interestingly hypoxia-exposed premyelinating oligodendrocytes (Mo3.13) showed reduced cholesterol along with decreased expression of HMGCR and SQLE on qPCR and Western blot. Exogenous cholesterol increased the differentiation of Mo3.13 as measured by increased expression of myelin basic protein (MBP) on flow cytometry. Hypoxia exposure resulted in increased cholesterol transport from astrocytes to oligodendrocytes in cocultures with BODIPY-cholesterol labelled SVG and membrane-labelled Mo3.13. As exogenous cholesterol enhanced oligodendrocyte differentiation, our findings indicate that increased cholesterol synthesis by astrocytes and transport to oligodendrocytes could supplement oligodendroglial maturation in conditions of hypoxic brain injury in neonates.
Collapse
Affiliation(s)
- Vadanya Shrivastava
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Sagar Tyagi
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Devanjan Dey
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Archna Singh
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | | | - Subrata Sinha
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - J B Sharma
- Department of Obstetrics and Gynecology, All India Institute of Medical Sciences, New Delhi, India
| | - Pankaj Seth
- Department of Molecular and Cellular Neuroscience, National Brain Research Centre, Manesar, Haryana, India
| | - Sudip Sen
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
42
|
Cases-Cunillera S, Quatraccioni A, Rossini L, Ruffolo G, Ono T, Baulac S, Auvin S, O'Brien TJ, Henshall DC, Akman Ö, Sankar R, Galanopoulou AS. WONOEP appraisal: The role of glial cells in focal malformations associated with early onset epilepsies. Epilepsia 2024; 65:3457-3468. [PMID: 39401070 DOI: 10.1111/epi.18126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 10/15/2024]
Abstract
Epilepsy represents a common neurological disorder in patients with developmental brain lesions, particularly in association with malformations of cortical development and low-grade glioneuronal tumors. In these diseases, genetic and molecular alterations in neurons are increasingly discovered that can trigger abnormalities in the neuronal network, leading to higher neuronal excitability levels. However, the mechanisms underlying epilepsy cannot rely solely on assessing the neuronal component. Growing evidence has revealed the high degree of complexity underlying epileptogenic processes, in which glial cells emerge as potential modulators of neuronal activity. Understanding the role of glial cells in developmental brain lesions such as malformations of cortical development and low-grade glioneuronal tumors is crucial due to the high degree of pharmacoresistance characteristic of these lesions. This has prompted research to investigate the role of glial and immune cells in epileptiform activity to find new therapeutic targets that could be used as combinatorial drug therapy. In a special session of the XVI Workshop of the Neurobiology of Epilepsy (WONOEP, Talloires, France, July 2022) organized by the Neurobiology Commission of the International League Against Epilepsy, we discussed the evidence exploring the genetic and molecular mechanisms of glial cells and immune response and their implications in the pathogenesis of neurodevelopmental pathologies associated with early life epilepsies.
Collapse
Affiliation(s)
- Silvia Cases-Cunillera
- Université Paris Cité, Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Neuronal Signaling in Epilepsy and Glioma, Paris, France
| | - Anne Quatraccioni
- Institute of Neuropathology, Section for Translational Epilepsy Research, Medical Faculty, University of Bonn, Bonn, Germany
| | - Laura Rossini
- Epilepsy Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Gabriele Ruffolo
- Department of Physiology and Pharmacology, Istituto Pasteur-Fondazione Cenci Bolognetti, University of Rome Sapienza, Rome, Italy
- IRCCS San Raffaele Roma, Rome, Italy
| | - Tomonori Ono
- Epilepsy Center, National Hospital Organization Nagasaki Medical Center, Ōmura, Japan
| | - Stéphanie Baulac
- Sorbonne Université, Institut du Cerveau-Paris Brain Institute-ICM, INSERM, CNRS, AP-HP, Hôpital de la Pitié Salpêtrière, Paris, France
| | - Stéphane Auvin
- Pediatric Neurology Department, AP-HP, Robert Debré University Hospital, CRMR épilepsies Rares, EpiCARE member, Paris, France
- Université Paris Cité, INSERM NeuroDiderot, Paris, France
- Institut Universitaire de France, Paris, France
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- Department of Neurology, Royal Melbourne Hospital, Melbourne, Victoria, Australia
- Department of Neurology, Alfred Health, Melbourne, Victoria, Australia
- Department of Medicine (Royal Melbourne Hospital), University of Melbourne, Melbourne, Victoria, Australia
| | - David C Henshall
- Department of Physiology and Medical Physics, RCSI, University of Medicine and Health Sciences, Dublin, Ireland
| | - Özlem Akman
- Department of Physiology, Faculty of Medicine, Demiroglu Bilim University, Istanbul, Turkey
| | - Raman Sankar
- Department of Pediatrics and Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Aristea S Galanopoulou
- Saul R. Korey Department of Neurology, Isabelle Rapin Division of Child Neurology, Dominique P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
43
|
Barbosa FMS, Dos Santos IR, de Almeida BA, Molossi FA, de Almeida PR, Lamego EC, Barth JC, Simões SVD, Panziera W, Sonne L, Pavarini SP, Driemeier D. Comparative study of non-suppurative meningoencephalitis in cattle in Southern Brazil. Vet Res Commun 2024; 48:4079-4088. [PMID: 39215894 DOI: 10.1007/s11259-024-10524-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Viral neurologic diseases are common in cattle, although most non-suppurative meningoencephalitis (NSM) remains etiologically unknown. We compared the epidemiological, clinical, and pathological data among 79 cases of rabies, 12 cases of NSM of unknown etiology (NSM-UE), and 8 cases of herpetic meningoencephalitis previously diagnosed in cattle in Southern Brazil. Neurological clinical signs were similar among rabies and NSM-UE and different in cattle with herpetic meningoencephalitis. Only two herpetic meningoencephalitis cases had gross lesions in the central nervous system, characterized by malacia and hemorrhage. Histologically, all three groups had mild to severe multifocal infiltrates of lymphocytes, plasma cells, and macrophages/microglial cells in the Virchow-Robin space, neuropil, and leptomeninges, and gliosis. Other findings included malacia and eosinophilic intracytoplasmic inclusion in rabies, and malacia and intranuclear amphophilic inclusion in herpetic meningoencephalitis. By immunohistochemistry, the predominant inflammatory cells in all cases were T lymphocytes, followed by macrophages/microglial cells, B lymphocytes, and astrocytes. The T lymphocyte count showed statistically significant differences between the diseases. Our results revealed few differences between the groups. Although the etiological agent involved has not been identified in cases of NSM-UE, the characteristics observed in this study showed similarity with viral diseases.
Collapse
Affiliation(s)
- Francisca Maria Sousa Barbosa
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil.
| | - Igor Ribeiro Dos Santos
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Bruno Albuquerque de Almeida
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Franciéli Adriane Molossi
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Eryca Ceolin Lamego
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Júlia Camargo Barth
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Welden Panziera
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Luciana Sonne
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Saulo Petinatti Pavarini
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - David Driemeier
- Setor de Patologia Veterinária, Faculdade de Veterinária, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
44
|
Feng H, Luo J, Li Z, Zhao Y, Liu Y, Zhu H. Valproic acid attenuates the severity of astrogliosis in the hippocampus of animal models of temporal lobe epilepsy. IBRO Neurosci Rep 2024; 17:471-479. [PMID: 39669223 PMCID: PMC11635005 DOI: 10.1016/j.ibneur.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 11/04/2024] [Indexed: 12/14/2024] Open
Abstract
Reactive astrogliosis is one of the most frequency neuropathological alterations in the hippocampus of animal models and patients with temporal lobe epilepsy (TLE). Valproic acid (VPA), a widely used antiepileptic drug (AED), acts by blocking ion channels and enhancing GABAergic activity. This study investigated the effects of VPA on hippocampal astrogliosis in a rat model of TLE. The results demonstrated that chronic administration of VPA at a dose of 200 mg/kg significantly reduced the severity of astrogliosis and ameliorated neuronal loss in the hippocampus at the early and middle stages post-status epilepticus (SE), while also improving cognitive impairments at the middle and late stages in KA-SE rats. Long-term administration of VPA at 400 mg/kg attenuated astrogliosis in the hippocampus at the middle stage post-SE, but lacked neuroprotective effects and exacerbated cognitive impairments at the late stage. These findings suggest that VPA at an appropriate dose could mitigate hippocampal astrogliosis, potentially offering a new antiepileptic mechanism for its long-term use.
Collapse
Affiliation(s)
- Hu Feng
- School of Life Sciences, Shanghai University, Nanchen Road 333, Shanghai 200436, China
| | - Jiamin Luo
- School of Life Sciences, Shanghai University, Nanchen Road 333, Shanghai 200436, China
| | - Zhiwei Li
- School of Life Sciences, Shanghai University, Nanchen Road 333, Shanghai 200436, China
| | - Yuxiao Zhao
- School of Life Sciences, Shanghai University, Nanchen Road 333, Shanghai 200436, China
| | - Yamei Liu
- School of Life Sciences, Shanghai University, Nanchen Road 333, Shanghai 200436, China
| | - Hongyan Zhu
- School of Life Sciences, Shanghai University, Nanchen Road 333, Shanghai 200436, China
| |
Collapse
|
45
|
González-Flores O, Garcia-Juárez M, Tecamachaltzi-Silvarán MB, Lucio RA, Ordoñez RD, Pfaus JG. Cellular and molecular mechanisms of action of ovarian steroid hormones. I: Regulation of central nervous system function. Neurosci Biobehav Rev 2024; 167:105937. [PMID: 39510217 DOI: 10.1016/j.neubiorev.2024.105937] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/25/2024] [Accepted: 10/31/2024] [Indexed: 11/15/2024]
Abstract
The conventional way steroid hormones work through receptors inside cells is widely acknowledged. There are unanswered questions about what happens to the hormone in the end and why there isn't always a strong connection between how much tissue takes up and its biological effects through receptor binding. Steroid hormones can also have non-traditional effects that happen quickly but don't involve entering the cell. Several possible mechanisms for these non-traditional actions include (a) changes in membrane fluidity, (b) steroid hormones acting on receptors on the outer surface of cells, (c) steroid hormones regulating GABAA receptors on cell membranes, and (d) activation of steroid receptors by factors like EGF, IGF-1, and dopamine. Data also suggests that steroid hormones may be inserted into DNA through receptors, acting as transcription factors. These proposed new mechanisms of action should not be seen as challenging the conventional mechanism. Instead, they contribute to a more comprehensive understanding of how hormones work, allowing for rapid, short-term, and prolonged effects to meet the body's physiological needs.
Collapse
Affiliation(s)
- Oscar González-Flores
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico.
| | - Marcos Garcia-Juárez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico
| | | | - Rosa Angélica Lucio
- Centro Tlaxcala de Biología de la Conducta, Universidad Autónoma de Tlaxcala, Tlaxcala, Mexico
| | - Raymundo Domínguez Ordoñez
- Centro de Investigación en Reproducción Animal, Universidad Autónoma de Tlaxcala-CINVESTAV, Tlaxcala, Mexico; Licenciatura en Ingeniería Agronómica y Zootecnia, Complejo Regional Centro, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - James G Pfaus
- Center for Sexual Health and Intervention, Czech National Institute of Mental Health, Klecany, Czech Republic; Department of Psychology and Life Sciences, Faculty of Humanities, Charles University, Prague, Czech Republic
| |
Collapse
|
46
|
Ye Y, Su X, Tang J, Zhu C. Neuropathic Pain Induced by Spinal Cord Injury from the Glia Perspective and Its Treatment. Cell Mol Neurobiol 2024; 44:81. [PMID: 39607514 PMCID: PMC11604677 DOI: 10.1007/s10571-024-01517-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Regional neuropathic pain syndromes above, at, or below the site of spinal damage arise after spinal cord injury (SCI) and are believed to entail distinct pathways; nevertheless, they may share shared defective glial systems. Neuropathic pain after SCI is caused by glial cells, ectopic firing of neurons endings and their intra- and extracellular signaling mechanisms. One such mechanism occurs when stimuli that were previously non-noxious become so after the injury. This will exhibit a symptom of allodynia. Another mechanism is the release of substances by glia, which keeps the sensitivity of dorsal horn neurons even in regions distant from the site of injury. Here, we review, the models and identifications of SCI-induced neuropathic pain (SCI-NP), the mechanisms of SCI-NP related to glia, and the treatments of SCI-NP.
Collapse
Affiliation(s)
- Ying Ye
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Anesthesiology, Jinling Hospital, Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinjin Su
- Department of Orthopedics, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Tang
- Department of Anesthesiology, Affiliated Hospital of Medical School, Jinling Hospital, Nanjing University, Nanjing, China
| | - Chao Zhu
- Department of Spine Surgery, School of Medicine, Renji Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
47
|
May HG, Tsikonofilos K, Donat CK, Sastre M, Kozlov AS, Sharp DJ, Bruyns-Haylett M. EEG hyperexcitability and hyperconnectivity linked to GABAergic inhibitory interneuron loss following traumatic brain injury. Brain Commun 2024; 6:fcae385. [PMID: 39605970 PMCID: PMC11600960 DOI: 10.1093/braincomms/fcae385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 09/04/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Traumatic brain injury represents a significant global health burden and has the highest prevalence among neurological disorders. Even mild traumatic brain injury can induce subtle, long-lasting changes that increase the risk of future neurodegeneration. Importantly, this can be challenging to detect through conventional neurological assessment. This underscores the need for more sensitive diagnostic tools, such as electroencephalography, to uncover opportunities for therapeutic intervention. Progress in the field has been hindered by a lack of studies linking mechanistic insights at the microscopic level from animal models to the macroscale phenotypes observed in clinical imaging. Our study addresses this gap by investigating a rat model of mild blast traumatic brain injury using both immunohistochemical staining of inhibitory interneurons and translationally relevant electroencephalography recordings. Although we observed no pronounced effects immediately post-injury, chronic time points revealed broadband hyperexcitability and increased connectivity, accompanied by decreased density of inhibitory interneurons. This pattern suggests a disruption in the balance between excitation and inhibition, providing a crucial link between cellular mechanisms and clinical hallmarks of injury. Our findings have significant implications for the diagnosis, monitoring, and treatment of traumatic brain injury. The emergence of electroencephalography abnormalities at chronic time points, despite the absence of immediate effects, highlights the importance of long-term monitoring in traumatic brain injury patients. The observed decrease in inhibitory interneuron density offers a potential cellular mechanism underlying the electroencephalography changes and may represent a target for therapeutic intervention. This study demonstrates the value of combining cellular-level analysis with macroscale neurophysiological recordings in animal models to elucidate the pathophysiology of traumatic brain injury. Future research should focus on translating these findings to human studies and exploring potential therapeutic strategies targeting the excitation-inhibition imbalance in traumatic brain injury.
Collapse
Affiliation(s)
- Hazel G May
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Konstantinos Tsikonofilos
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Department of Neuroscience, Karolinska Institutet, Stockholm 171 65, Sweden
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm 171 65, Sweden
| | - Cornelius K Donat
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
- Department of Medicinal Radiochemistry, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, 01328 Dresden, Germany
| | - Magdalena Sastre
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Andriy S Kozlov
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - David J Sharp
- Department of Brain Sciences, Imperial College London, London W12 0NN, UK
| | - Michael Bruyns-Haylett
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
- Department of Bioengineering, Institut Quimic de Sarria, Universitat Ramon Llul, Barcelona 08017, Spain
- Department of Quantitative Methods, Institut Quimic de Sarria, Universitat Ramon Llul, Barcelona 08017, Spain
| |
Collapse
|
48
|
Ryu JE, Shim KW, Roh HW, Park M, Lee JH, Kim EY. Circadian regulation of endoplasmic reticulum calcium response in cultured mouse astrocytes. eLife 2024; 13:RP96357. [PMID: 39601391 PMCID: PMC11602189 DOI: 10.7554/elife.96357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2024] Open
Abstract
The circadian clock, an internal time-keeping system orchestrates 24 hr rhythms in physiology and behavior by regulating rhythmic transcription in cells. Astrocytes, the most abundant glial cells, play crucial roles in CNS functions, but the impact of the circadian clock on astrocyte functions remains largely unexplored. In this study, we identified 412 circadian rhythmic transcripts in cultured mouse cortical astrocytes through RNA sequencing. Gene Ontology analysis indicated that genes involved in Ca2+ homeostasis are under circadian control. Notably, Herpud1 (Herp) exhibited robust circadian rhythmicity at both mRNA and protein levels, a rhythm disrupted in astrocytes lacking the circadian transcription factor, BMAL1. HERP regulated endoplasmic reticulum (ER) Ca2+ release by modulating the degradation of inositol 1,4,5-trisphosphate receptors (ITPRs). ATP-stimulated ER Ca2+ release varied with the circadian phase, being more pronounced at subjective night phase, likely due to the rhythmic expression of ITPR2. Correspondingly, ATP-stimulated cytosolic Ca2+ increases were heightened at the subjective night phase. This rhythmic ER Ca2+ response led to circadian phase-dependent variations in the phosphorylation of Connexin 43 (Ser368) and gap junctional communication. Given the role of gap junction channel (GJC) in propagating Ca2+ signals, we suggest that this circadian regulation of ER Ca2+ responses could affect astrocytic modulation of synaptic activity according to the time of day. Overall, our study enhances the understanding of how the circadian clock influences astrocyte function in the CNS, shedding light on their potential role in daily variations of brain activity and health.
Collapse
Affiliation(s)
- Ji Eun Ryu
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Brain Science, Ajou University School of MedicineSuwonRepublic of Korea
| | - Kyu-Won Shim
- Interdisciplinary Program in Bioinformatics, Seoul National UniversitySeoulRepublic of Korea
| | - Hyun Woong Roh
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Psychiatry, Ajou University School of MedicineSuwonRepublic of Korea
| | - Minsung Park
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Brain Science, Ajou University School of MedicineSuwonRepublic of Korea
| | - Jae-Hyung Lee
- Department of Oral Microbiology, College of Dentistry, Kyung Hee UniversitySeoulRepublic of Korea
| | - Eun Young Kim
- Neuroscience Graduate Program, Department of Biomedical Sciences, Ajou University Graduate School of MedicineSuwonRepublic of Korea
- Department of Brain Science, Ajou University School of MedicineSuwonRepublic of Korea
| |
Collapse
|
49
|
Liu H, Tan AYS, Mehrabi NF, Turner CP, Curtis MA, Faull RLM, Dragunow M, Singh-Bains MK, Smith AM. Astrocytic proteins involved in regulation of the extracellular environment are increased in the Alzheimer's disease middle temporal gyrus. Neurobiol Dis 2024; 204:106749. [PMID: 39603277 DOI: 10.1016/j.nbd.2024.106749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
Alzheimer's disease (AD) has complex pathophysiology involving numerous cell types and brain processes. Astrocyte involvement in AD is gaining increased attention, however a complete characterisation of astrocytic changes in the AD human brain is warranted. Astrocytes perform important homeostatic functions including regulation of the extracellular microenvironment, critical for the health of all brain cells. We have investigated changes to key astrocyte proteins involved in the regulation of CNS extracellular environment in the human AD middle temporal gyrus (MTG): aquaporin-4 (AQP-4), glutamate transporter-1 (GLT-1) and inwardly-rectifying potassium channel 4.1 (Kir4.1). We have used a high-throughput human brain tissue microarray platform with automated quantitative image analysis to measure protein changes in a large cohort of neurological control and AD cases. We found increased astrocytic glial acidic fibrillary protein (GFAP), AQP-4, GLT-1 and Kir4.1 expression that correlates with advancing Braak stage, increasing amyloid pathology and, to a greater extent, the degree of tau pathology. We confirmed that Kir4.1 immunostaining is predominantly found in astrocytes and revealed a novel redistribution of Kir4.1 protein expression into astrocytic processes in the AD MTG. Our study presents novel and potentially modifiable glial changes in the AD human brain that are critical to our understanding of disease pathogenesis.
Collapse
Affiliation(s)
- Henry Liu
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Adelie Y S Tan
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Nasim F Mehrabi
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Clinton P Turner
- Department of Anatomical Pathology, Pathology and Laboratory Medicine, Auckland City Hospital, Auckland, New Zealand
| | - Maurice A Curtis
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Richard L M Faull
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Mike Dragunow
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand
| | - Malvindar K Singh-Bains
- Centre for Brain Research and Department of Anatomy and Medical Imaging, University of Auckland, Auckland, New Zealand
| | - Amy M Smith
- Centre for Brain Research and Department of Pharmacology and Clinical Pharmacology, University of Auckland, New Zealand.
| |
Collapse
|
50
|
Wu J, Tang J, Huang D, Wang Y, Zhou E, Ru Q, Xu G, Chen L, Wu Y. Effects and mechanisms of APP and its cleavage product Aβ in the comorbidity of sarcopenia and Alzheimer's disease. Front Aging Neurosci 2024; 16:1482947. [PMID: 39654807 PMCID: PMC11625754 DOI: 10.3389/fnagi.2024.1482947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/11/2024] [Indexed: 12/12/2024] Open
Abstract
Sarcopenia and AD are both classic degenerative diseases, and there is growing epidemiological evidence of their comorbidity with aging; however, the mechanisms underlying the biology of their commonality have not yet been thoroughly investigated. APP is a membrane protein that is expressed in tissues and is expressed not only in the nervous system but also in the NMJ and muscle. Deposition of its proteolytic cleavage product, Aβ, has been described as a central component of AD pathogenesis. Recent studies have shown that excessive accumulation and aberrant expression of APP in muscle lead to pathological muscle lesions, but the pathogenic mechanism by which APP and its proteolytic cleavage products act in skeletal muscle is less well understood. By summarizing and analyzing the literature concerning the role, pathogenicity and pathological mechanisms of APP and its cleavage products in the nervous system and muscles, we aimed to explore the intrinsic pathological mechanisms of myocerebral comorbidities and to provide new perspectives and theoretical foundations for the prevention and treatment of AD and sarcopenia comorbidities.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lin Chen
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| | - Yuxiang Wu
- Institute of Intelligent Sport and Proactive Health, Department of Health and Physical Education, Jianghan University, Wuhan, China
| |
Collapse
|