1
|
Anwar S, Zhou J, Kowalski L, Saylor J, Shukla D, Boetel K, Song Z, Sharma K, Cheng J, Inoue M. Star-Shaped Glatiramer Acetate Mitigates Pulmonary Dysfunction and Brain Neurodegeneration in a Murine Model of Cryptococcus-Associated IRIS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.07.631707. [PMID: 39829780 PMCID: PMC11741261 DOI: 10.1101/2025.01.07.631707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2025]
Abstract
Cryptococcal-associated immune reconstitution inflammatory syndrome (C-IRIS) is a clinical worsening or new presentation of cryptococcal disease following the initiation of antiretroviral therapy. C-IRIS is primarily driven by an influx of pathological CD4 + T cells, which triggers a hyperinflammatory response. The murine model of C-IRIS is a way to study the disease in mice and understand how the immune system triggers life-threatening outcomes in patients. We previously developed a murine C-IRIS model and demonstrated that C-IRIS is triggered by pathological CD4 + T cells, particularly Th1 cells, in the brain, which triggers neurodegeneration and pulmonary dysfunction. Using this unique mouse model, we tested the therapeutic effect of a star-shaped glatiramer acetate (sGA), which is a more effective isomeric form than linear GA. Here, we observed that sGA suppresses Th1 differentiation in the lung tissues, reducing CD4 + T cell and Th1 cell count. It also reduced microglia populations in the brain. Together, these changes improved respiratory dysfunction caused by C-IRIS, lowered mortality rate, and reduced brain neurodegeneration. These findings suggest that sGA could be an effective therapeutic strategy for managing C-IRIS.
Collapse
|
2
|
Li M, Chen M, Li H, Gao D, Zhao L, Zhu M. Glial cells improve Parkinson's disease by modulating neuronal function and regulating neuronal ferroptosis. Front Cell Dev Biol 2025; 12:1510897. [PMID: 39830208 PMCID: PMC11739109 DOI: 10.3389/fcell.2024.1510897] [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: 10/14/2024] [Accepted: 12/11/2024] [Indexed: 01/22/2025] Open
Abstract
The main characteristics of Parkinson's disease (PD) are the loss of dopaminergic (DA) neurons and abnormal aggregation of cytosolic proteins. However, the exact pathogenesis of PD remains unclear, with ferroptosis emerging as one of the key factors driven by iron accumulation and lipid peroxidation. Glial cells, including microglia, astrocytes, and oligodendrocytes, serve as supportive cells in the central nervous system (CNS), but their abnormal activation can lead to DA neuron death and ferroptosis. This paper explores the interactions between glial cells and DA neurons, reviews the changes in glial cells during the pathological process of PD, and reports on how glial cells regulate ferroptosis in PD through iron homeostasis and lipid peroxidation. This opens up a new pathway for basic research and therapeutic strategies in Parkinson's disease.
Collapse
Affiliation(s)
- Mengzhu Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Mengxuan Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Haiyan Li
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| | - Da Gao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Lijun Zhao
- Shenzhen Clinical College of Integrated Chinese and Western Medicine, Guangzhou University of Chinese Medicine, Shenzhen, China
| | - Meiling Zhu
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China
| |
Collapse
|
3
|
Al‐kuraishy HM, Sulaiman GM, Mohammed HA, Albukhaty S, Albuhadily AK, Al‐Gareeb AI, Klionsky DJ, Abomughaid MM. The Compelling Role of Brain-Derived Neurotrophic Factor Signaling in Multiple Sclerosis: Role of BDNF Activators. CNS Neurosci Ther 2024; 30:e70167. [PMID: 39654365 PMCID: PMC11628746 DOI: 10.1111/cns.70167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 11/14/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024] Open
Abstract
Brain-derived neurotrophic factor (BDNF) is a neurotrophin, acting as a neurotrophic signal and neuromodulator in the central nervous system (CNS). BDNF is synthesized from its precursor proBDNF within the CNS and peripheral tissues. Through activation of NTRK2/TRKB (neurotrophic receptor tyrosine kinase 2), BDNF promotes neuronal survival, synaptic plasticity, and neuronal growth, whereas it inhibits microglial activation and the release of pro-inflammatory cytokines. BDNF is dysregulated in different neurodegenerative diseases and depressions. However, there is a major controversy concerning BDNF levels in the different stages of multiple sclerosis (MS). Therefore, this review discusses the potential role of BDNF signaling in stages of MS, and how BDNF modulators affect the pathogenesis and outcomes of this disease.
Collapse
Affiliation(s)
- Hayder M. Al‐kuraishy
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | - Ghassan M. Sulaiman
- Division of Biotechnology, Department of Applied SciencesUniversity of TechnologyBaghdadIraq
| | - Hamdoon A. Mohammed
- Department of Medicinal Chemistry and Pharmacognosy, College of PharmacyQassim UniversityQassimSaudi Arabia
| | | | - Ali K. Albuhadily
- Department of Clinical Pharmacology and Medicine, College of MedicineMustansiriyah UniversityBaghdadIraq
| | | | | | - Mosleh M. Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical SciencesUniversity of BishaBishaSaudi Arabia
| |
Collapse
|
4
|
Woodfin S, Hall S, Ramerth A, Chapple B, Fausnacht D, Moore W, Alkhalidy H, Liu D. Potential Application of Plant-Derived Compounds in Multiple Sclerosis Management. Nutrients 2024; 16:2996. [PMID: 39275311 PMCID: PMC11397714 DOI: 10.3390/nu16172996] [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: 07/30/2024] [Revised: 08/23/2024] [Accepted: 08/29/2024] [Indexed: 09/16/2024] Open
Abstract
Multiple sclerosis (MS) is a chronic autoimmune disorder characterized by inflammation, demyelination, and neurodegeneration, resulting in significant disability and reduced quality of life. Current therapeutic strategies primarily target immune dysregulation, but limitations in efficacy and tolerability highlight the need for alternative treatments. Plant-derived compounds, including alkaloids, phenylpropanoids, and terpenoids, have demonstrated anti-inflammatory effects in both preclinical and clinical studies. By modulating immune responses and promoting neuroregeneration, these compounds offer potential as novel adjunctive therapies for MS. This review provides insights into the molecular and cellular basis of MS pathogenesis, emphasizing the role of inflammation in disease progression. It critically evaluates emerging evidence supporting the use of plant-derived compounds to attenuate inflammation and MS symptomology. In addition, we provide a comprehensive source of information detailing the known mechanisms of action and assessing the clinical potential of plant-derived compounds in the context of MS pathogenesis, with a focus on their anti-inflammatory and neuroprotective properties.
Collapse
Affiliation(s)
- Seth Woodfin
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Sierra Hall
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Alexis Ramerth
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Brooke Chapple
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Dane Fausnacht
- Department of Biology, School of Sciences and Agriculture, Ferrum College, Ferrum, VA 24088, USA
| | - William Moore
- Department of Biology and Chemistry, School of Health Sciences, Liberty University, Lynchburg, VA 24515, USA
| | - Hana Alkhalidy
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
- Department of Nutrition and Food Technology, Faculty of Agriculture, Jordan University of Science and Technology, P.O. Box 3030, Irbid 22110, Jordan
| | - Dongmin Liu
- Department of Human Nutrition, Foods and Exercise, College of Agriculture and Life Sciences, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
5
|
Gaire BP, Koronyo Y, Fuchs DT, Shi H, Rentsendorj A, Danziger R, Vit JP, Mirzaei N, Doustar J, Sheyn J, Hampel H, Vergallo A, Davis MR, Jallow O, Baldacci F, Verdooner SR, Barron E, Mirzaei M, Gupta VK, Graham SL, Tayebi M, Carare RO, Sadun AA, Miller CA, Dumitrascu OM, Lahiri S, Gao L, Black KL, Koronyo-Hamaoui M. Alzheimer's disease pathophysiology in the Retina. Prog Retin Eye Res 2024; 101:101273. [PMID: 38759947 PMCID: PMC11285518 DOI: 10.1016/j.preteyeres.2024.101273] [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: 02/11/2023] [Revised: 04/23/2024] [Accepted: 05/10/2024] [Indexed: 05/19/2024]
Abstract
The retina is an emerging CNS target for potential noninvasive diagnosis and tracking of Alzheimer's disease (AD). Studies have identified the pathological hallmarks of AD, including amyloid β-protein (Aβ) deposits and abnormal tau protein isoforms, in the retinas of AD patients and animal models. Moreover, structural and functional vascular abnormalities such as reduced blood flow, vascular Aβ deposition, and blood-retinal barrier damage, along with inflammation and neurodegeneration, have been described in retinas of patients with mild cognitive impairment and AD dementia. Histological, biochemical, and clinical studies have demonstrated that the nature and severity of AD pathologies in the retina and brain correspond. Proteomics analysis revealed a similar pattern of dysregulated proteins and biological pathways in the retina and brain of AD patients, with enhanced inflammatory and neurodegenerative processes, impaired oxidative-phosphorylation, and mitochondrial dysfunction. Notably, investigational imaging technologies can now detect AD-specific amyloid deposits, as well as vasculopathy and neurodegeneration in the retina of living AD patients, suggesting alterations at different disease stages and links to brain pathology. Current and exploratory ophthalmic imaging modalities, such as optical coherence tomography (OCT), OCT-angiography, confocal scanning laser ophthalmoscopy, and hyperspectral imaging, may offer promise in the clinical assessment of AD. However, further research is needed to deepen our understanding of AD's impact on the retina and its progression. To advance this field, future studies require replication in larger and diverse cohorts with confirmed AD biomarkers and standardized retinal imaging techniques. This will validate potential retinal biomarkers for AD, aiding in early screening and monitoring.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ron Danziger
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jean-Philippe Vit
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Nazanin Mirzaei
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jonah Doustar
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Harald Hampel
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Andrea Vergallo
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France
| | - Miyah R Davis
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ousman Jallow
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Filippo Baldacci
- Sorbonne University, Alzheimer Precision Medicine (APM), AP-HP, Pitié-Salpêtrière Hospital, Paris, France; Department of Clinical and Experimental Medicine, Neurology Unit, University of Pisa, Pisa, Italy
| | | | - Ernesto Barron
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Vivek K Gupta
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia
| | - Stuart L Graham
- Department of Clinical Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, Australia; Department of Clinical Medicine, Macquarie University, Sydney, NSW, Australia
| | - Mourad Tayebi
- School of Medicine, Western Sydney University, Campbelltown, NSW, Australia
| | - Roxana O Carare
- Department of Clinical Neuroanatomy, University of Southampton, Southampton, UK
| | - Alfredo A Sadun
- Department of Ophthalmology, David Geffen School of Medicine at University of California Los Angeles, Los Angeles, CA, USA; Doheny Eye Institute, Los Angeles, CA, USA
| | - Carol A Miller
- Department of Pathology Program in Neuroscience, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | | | - Shouri Lahiri
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Liang Gao
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, USA
| | - Keith L Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, USA; Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
6
|
Gil-Sánchez A, Gonzalo H, Canudes M, Nogueras L, González-Mingot C, Valcheva P, Torres P, Serrano JC, Peralta S, Solana MJ, Brieva L. Can Glatiramer Acetate Prevent Cognitive Impairment by Modulating Oxidative Stress in Patients with Multiple Sclerosis? Pharmaceuticals (Basel) 2024; 17:459. [PMID: 38675419 PMCID: PMC11053874 DOI: 10.3390/ph17040459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disease characterized by demyelination and neuroinflammation, often accompanied by cognitive impairment. This study aims (1) to investigate the potential of glatiramer acetate (GA) as a therapy for preventing cognitive decline in patients with MS (pwMS) by modulating oxidative stress (OS) and (2) to seek out the differences in cognition between pwMS in a cohort exhibiting good clinical evolution and control subjects (CS). An exploratory, prospective, multicentre, cross-sectional case-control study was conducted, involving three groups at a 1:1:1 ratio-41 GA-treated pwMS, 42 untreated pwMS, and 42 CS. The participants performed a neuropsychological battery and underwent venepuncture for blood sampling. The inclusion criteria required an Expanded Disability Status Scale score of ≤3.0 and a minimum of 5 years of MS disease. Concerning cognition, the CS had a better performance than the pwMS (p = <0.0001), and between those treated and untreated with GA, no statistically significant differences were found. Regarding oxidation, no statistically significant differences were detected. Upon categorizing the pwMS into cognitively impaired and cognitively preserved groups, the lactate was elevated in the pwMS with cognitive preservation (p = 0.038). The pwMS exhibited a worse cognitive performance than the CS. The pwMS treated with GA did not show an improvement in oxidation. Lactate emerged as a potential biomarker for cognitive preservation.
Collapse
Affiliation(s)
- Anna Gil-Sánchez
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
| | - Hugo Gonzalo
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
| | - Marc Canudes
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
| | - Lara Nogueras
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
| | - Cristina González-Mingot
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
- Hospital Universitario Arnau de Vilanova de Lleida (HUAVLleida), 25198 Lleida, Spain
- Neuroimmunology Group, Department of Medicine, University of Lleida, 25198 Lleida, Spain;
| | - Petya Valcheva
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
| | - Pascual Torres
- Neuroimmunology Group, Department of Medicine, University of Lleida, 25198 Lleida, Spain;
| | - Jose Carlos Serrano
- NUTREN-Nutrigenomics, Department of Experimental Medicine, University of Lleida, 25198 Lleida, Spain;
| | - Silvia Peralta
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
- Hospital Universitario Arnau de Vilanova de Lleida (HUAVLleida), 25198 Lleida, Spain
| | - Maria José Solana
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
- Hospital Universitario Arnau de Vilanova de Lleida (HUAVLleida), 25198 Lleida, Spain
| | - Luis Brieva
- Institut de Recerca Biomèdica de Lleida (IRBLleida), 25198 Lleida, Spain; (H.G.); (M.C.); (L.N.); (C.G.-M.); (P.V.); (S.P.); (M.J.S.)
- Hospital Universitario Arnau de Vilanova de Lleida (HUAVLleida), 25198 Lleida, Spain
- Neuroimmunology Group, Department of Medicine, University of Lleida, 25198 Lleida, Spain;
| |
Collapse
|
7
|
Huang Z, Gong Z, Lin Y, Yang F, Chen W, Xiang S, Huang Y, Xiao H, Xu S, Duan J. Treatment with glatiramer acetate in APPswe/PS1dE9 mice at an early stage of Alzheimer's disease prior to amyloid-beta deposition delays the disease's pathological development and ameliorates cognitive decline. Front Aging Neurosci 2024; 16:1267780. [PMID: 38352237 PMCID: PMC10861656 DOI: 10.3389/fnagi.2024.1267780] [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: 07/27/2023] [Accepted: 01/11/2024] [Indexed: 02/16/2024] Open
Abstract
Background Alzheimer's disease (AD) is characterized by neuroinflammation, which is frequently accompanied by immune system dysfunction. Although the mechanism of neurodegenerative lesions is unclear, various clinical trials have highlighted that early intervention in AD is crucial to the success of treatment. In order to explore the potential of immunotherapy in the early period of AD, the present study evaluated whether application of glatiramer acetate (GA), an immunomodulatory agent approved for remitting-relapsing multiple sclerosis (RRMS), in the early stages of AD prior to amyloid beta (Aβ) deposition altered the Aβ pathology and cognitive impairments in APPswe/PSEN1dE9 (APP/PS1) transgenic mice. Methods We treated two cohorts of pre-depositing and amyloid-depositing (2- and 6-month-old) APP/PS1 mice with weekly-GA subcutaneous injection over a 12-week period. We then tested spatial learning and memory using the Morris water maze (MWM) and the Y maze. Immunohistochemistry staining was utilized to analyze Aβ burden in the brain as well as activated microglia. Furthermore, the inflammatory cytokine milieu within brains was estimated by quantitative real-time polymerase chain reaction, and the peripheral CD4+CD25+Foxp3+ regulatory T cells (Tregs) in the spleen were measured by flow cytometry. Results We found that early GA administration reduced Aβ burden and ameliorated cognitive decline. Meanwhile, the immune microenvironment had changed in the brain, with an increase in the production of anti-inflammatory cytokines and a decrease in microglial activation. Interestingly, early GA administration also modulated the peripheral immune system through the amplification of Tregs in the spleen. Conclusion Overall, our findings revealed that GA treatment might enhance the central and peripheral immune systems' protective capabilities in the early stages of AD, eventually improving cognitive deficits. Our research supports the advantages of immunomodulatory treatments for AD at an early stage.
Collapse
Affiliation(s)
- Zengyong Huang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
- Shantou Central Hospital, Shantou, China
| | - Zhuo Gong
- Shantou Central Hospital, Shantou, China
| | - Yongtai Lin
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Fan Yang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Weiping Chen
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Shaotong Xiang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Yuedong Huang
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Hao Xiao
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Shuwen Xu
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| | - Jinhai Duan
- Eastern Department of Neurology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Guangdong Geriatrics Institute and Guangdong Cardiovascular Institute, Southern Medical University, Guangzhou, China
| |
Collapse
|
8
|
Dejbakht M, Akhzari M, Jalili S, Faraji F, Barazesh M. Multiple Sclerosis: New Insights into Molecular Pathogenesis and Novel Platforms for Disease Treatment. Curr Drug Res Rev 2024; 16:175-197. [PMID: 37724675 DOI: 10.2174/2589977516666230915103730] [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: 01/08/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS), a chronic inflammatory disorder, affects the central nervous system via myelin degradation. The cause of MS is not fully known, but during recent years, our knowledge has deepened significantly regarding the different aspects of MS, including etiology, molecular pathophysiology, diagnosis and therapeutic options. Myelin basic protein (MBP) is the main myelin protein that accounts for maintaining the stability of the myelin sheath. Recent evidence has revealed that MBP citrullination or deamination, which is catalyzed by Ca2+ dependent peptidyl arginine deiminase (PAD) enzyme leads to the reduction of positive charge, and subsequently proteolytic cleavage of MBP. The overexpression of PAD2 in the brains of MS patients plays an essential role in new epitope formation and progression of the autoimmune disorder. Some drugs have recently entered phase III clinical trials with promising efficacy and will probably obtain approval in the near future. As different therapeutic platforms develop, finding an optimal treatment for each individual patient will be more challenging. AIMS This review provides a comprehensive insight into MS with a focus on its pathogenesis and recent advances in diagnostic methods and its present and upcoming treatment modalities. CONCLUSION MS therapy alters quickly as research findings and therapeutic options surrounding MS expand. McDonald's guidelines have created different criteria for MS diagnosis. In recent years, ever-growing interest in the development of PAD inhibitors has led to the generation of many reversible and irreversible PAD inhibitors against the disease with satisfactory therapeutic outcomes.
Collapse
Affiliation(s)
- Majid Dejbakht
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Fouziyeh Faraji
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- Department of Biotechnology, Cellular and Molecular Research Center, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
9
|
Rahiman N, Zamani P, Arabi L, Alavizadeh SH, Nikpoor A, Mashreghi M, Badiee A, Jaafari MR. Novel liposomal glatiramer acetate: Preparation and immunomodulatory evaluation in murine model of multiple sclerosis. Int J Pharm 2023; 648:123620. [PMID: 37981250 DOI: 10.1016/j.ijpharm.2023.123620] [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: 06/12/2023] [Revised: 10/23/2023] [Accepted: 11/15/2023] [Indexed: 11/21/2023]
Abstract
The frequent administration rate required for Glatiramer acetate (GA), a first-line therapy for Multiple sclerosis (MS), poses patient compliance issues. Only a small portion of the subcutaneously administered GA is available for phagocytosis by macrophages, as most of it is hydrolyzed at its administration site or excreted renally. To unravel these hurdles, we have prepared liposomal formulations of GA through thin film-hydration method plus extrusion. The clinical and histopathological efficacy of GA-loaded liposomes were assessed in prophylactic and therapeutic manners on murine model of MS (experimental autoimmune encephalomyelitis (EAE)). The selected GA liposomal formulation showed favorable size (275 nm on average), high loading efficiency, and high macrophage localization. Moreover, administration of GA-liposomes in mice robustly suppressed the inflammatory responses and decreased the inflammatory and demyelinated lesion regions in CNS compared to the free GA with subsequent reduction of the EAE clinical score. Our study indicated that liposomal GA could be served as a reliable nanomedicine-based platform to hopefully curb MS-related aberrant autoreactive immune responses with higher efficacy, longer duration of action, fewer administration frequencies, and higher delivery rate to macrophages. This platform has the potential to be introduced as a vaccine for MS after clinical translation and merits further investigations.
Collapse
Affiliation(s)
- Niloufar Rahiman
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Parvin Zamani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Leila Arabi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyedeh Hoda Alavizadeh
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Aminreza Nikpoor
- Molecular Medicine Research Center, Hormozgan Health Institute, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mohammad Mashreghi
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Badiee
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahmoud Reza Jaafari
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Pharmaceutical Nanotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
10
|
Zhai D, Yan S, Samsom J, Wang L, Su P, Jiang A, Zhang H, Jia Z, Wallach I, Heifets A, Zanato C, Tseng CC, Wong AH, Greig IR, Liu F. Small-molecule targeting AMPA-mediated excitotoxicity has therapeutic effects in mouse models for multiple sclerosis. SCIENCE ADVANCES 2023; 9:eadj6187. [PMID: 38064562 PMCID: PMC10708182 DOI: 10.1126/sciadv.adj6187] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/09/2023] [Indexed: 12/18/2023]
Abstract
While most research and treatments for multiple sclerosis (MS) focus on autoimmune reactions causing demyelination, it is possible that neurodegeneration precedes the autoimmune response. Hence, glutamate receptor antagonists preventing excitotoxicity showed promise in MS animal models, though blocking glutamate signaling prevents critical neuronal functions. This study reports the discovery of a small molecule that prevents AMPA-mediated excitotoxicity by targeting an allosteric binding site. A machine learning approach was used to screen for small molecules targeting the AMPA receptor GluA2 subunit. The lead candidate has potent effects in restoring neurological function and myelination while reducing the immune response in experimental autoimmune encephalitis and cuprizone MS mouse models without affecting basal neurotransmission or learning and memory. These findings facilitate development of a treatment for MS with a different mechanism of action than current immune modulatory drugs and avoids important off-target effects of glutamate receptor antagonists. This class of MS therapeutics could be useful as an alternative or complementary treatment to existing therapies.
Collapse
Affiliation(s)
- Dongxu Zhai
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
| | - Shuxin Yan
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
| | - James Samsom
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
| | - Le Wang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
| | - Ping Su
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
| | - Anlong Jiang
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
| | - Haorui Zhang
- Department of Neurosciences & Mental Health, The Hospital for Sick Children, 686 Bay St., Toronto M5G 0A4, Canada
| | - Zhengping Jia
- Department of Neurosciences & Mental Health, The Hospital for Sick Children, 686 Bay St., Toronto M5G 0A4, Canada
| | - Izhar Wallach
- Atomwise Inc., 221 Main Street, Suite 1350, San Francisco, CA 94105, USA
| | - Abraham Heifets
- Atomwise Inc., 221 Main Street, Suite 1350, San Francisco, CA 94105, USA
| | - Chiara Zanato
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Chih-Chung Tseng
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Albert H.C. Wong
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
- Institutes of Medical Science, University of Toronto, 1 King’s College Cir., Toronto M5S 1A8, Canada
- Department of Pharmacology and Toxicology, University of Toronto, 1 King’s College Cir., Toronto M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto M5T 1R8, Canada
| | - Iain R. Greig
- Institute of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen AB25 2ZD, Scotland, UK
| | - Fang Liu
- Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, 250 College St., Toronto M5T 1R8, Canada
- Institutes of Medical Science, University of Toronto, 1 King’s College Cir., Toronto M5S 1A8, Canada
- Department of Psychiatry, University of Toronto, 250 College St., Toronto M5T 1R8, Canada
- Department of Physiology, University of Toronto, 1 King’s College Cir., Toronto M5T 1R8, Canada
| |
Collapse
|
11
|
Mwema A, Muccioli GG, des Rieux A. Innovative drug delivery strategies to the CNS for the treatment of multiple sclerosis. J Control Release 2023; 364:435-457. [PMID: 37926243 DOI: 10.1016/j.jconrel.2023.10.052] [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: 07/05/2023] [Revised: 10/05/2023] [Accepted: 10/31/2023] [Indexed: 11/07/2023]
Abstract
Disorders of the central nervous system (CNS), such as multiple sclerosis (MS) represent a great emotional, financial and social burden. Despite intense efforts, great unmet medical needs remain in that field. MS is an autoimmune, chronic inflammatory demyelinating disease with no curative treatment up to date. The current therapies mostly act in the periphery and seek to modulate aberrant immune responses as well as slow down the progression of the disease. Some of these therapies are associated with adverse effects related partly to their administration route and show some limitations due to their rapid clearance and inability to reach the CNS. The scientific community have recently focused their research on developing MS therapies targeting different processes within the CNS. However, delivery of therapeutics to the CNS is mainly limited by the presence of the blood-brain barrier (BBB). Therefore, there is a pressing need to develop new drug delivery strategies that ensure CNS availability to capitalize on identified therapeutic targets. Several approaches have been developed to overcome or bypass the BBB and increase delivery of therapeutics to the CNS. Among these strategies, the use of alternative routes of administration, such as the nose-to-brain (N2B) pathway, offers a promising non-invasive option in the scope of MS, as it would allow a direct transport of the drugs from the nasal cavity to the brain. Moreover, the combination of bioactive molecules within nanocarriers bring forth new opportunities for MS therapies, allowing and/or increasing their transport to the CNS. Here we will review and discuss these alternative administration routes as well as the nanocarrier approaches useful to deliver drugs for MS.
Collapse
Affiliation(s)
- Ariane Mwema
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium; Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium
| | - Giulio G Muccioli
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Bioanalysis and Pharmacology of Bioactive Lipids, Avenue E. Mounier 72, 1200 Brussels, Belgium.
| | - Anne des Rieux
- Université catholique de Louvain, UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue E. Mounier 73, 1200 Brussels, Belgium.
| |
Collapse
|
12
|
Cáceres C, Heusser B, Garnham A, Moczko E. The Major Hypotheses of Alzheimer's Disease: Related Nanotechnology-Based Approaches for Its Diagnosis and Treatment. Cells 2023; 12:2669. [PMID: 38067098 PMCID: PMC10705786 DOI: 10.3390/cells12232669] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/25/2023] [Accepted: 09/19/2023] [Indexed: 12/18/2023] Open
Abstract
Alzheimer's disease (AD) is a well-known chronic neurodegenerative disorder that leads to the progressive death of brain cells, resulting in memory loss and the loss of other critical body functions. In March 2019, one of the major pharmaceutical companies and its partners announced that currently, there is no drug to cure AD, and all clinical trials of the new ones have been cancelled, leaving many people without hope. However, despite the clear message and startling reality, the research continued. Finally, in the last two years, the Food and Drug Administration (FDA) approved the first-ever medications to treat Alzheimer's, aducanumab and lecanemab. Despite researchers' support of this decision, there are serious concerns about their effectiveness and safety. The validation of aducanumab by the Centers for Medicare and Medicaid Services is still pending, and lecanemab was authorized without considering data from the phase III trials. Furthermore, numerous reports suggest that patients have died when undergoing extended treatment. While there is evidence that aducanumab and lecanemab may provide some relief to those suffering from AD, their impact remains a topic of ongoing research and debate within the medical community. The fact is that even though there are considerable efforts regarding pharmacological treatment, no definitive cure for AD has been found yet. Nevertheless, it is strongly believed that modern nanotechnology holds promising solutions and effective clinical strategies for the development of diagnostic tools and treatments for AD. This review summarizes the major hallmarks of AD, its etiological mechanisms, and challenges. It explores existing diagnostic and therapeutic methods and the potential of nanotechnology-based approaches for recognizing and monitoring patients at risk of irreversible neuronal degeneration. Overall, it provides a broad overview for those interested in the evolving areas of clinical neuroscience, AD, and related nanotechnology. With further research and development, nanotechnology-based approaches may offer new solutions and hope for millions of people affected by this devastating disease.
Collapse
Affiliation(s)
| | | | | | - Ewa Moczko
- Facultad de Ingeniería y Ciencias, Universidad Adolfo Ibáñez, Viña del Mar 2562307, Chile; (C.C.)
| |
Collapse
|
13
|
Tanelian A, Nankova B, Hu F, Sahawneh JD, Sabban EL. Effect of acetate supplementation on traumatic stress-induced behavioral impairments in male rats. Neurobiol Stress 2023; 27:100572. [PMID: 37781563 PMCID: PMC10539924 DOI: 10.1016/j.ynstr.2023.100572] [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: 06/28/2023] [Revised: 09/06/2023] [Accepted: 09/16/2023] [Indexed: 10/03/2023] Open
Abstract
Gut microbiota and their metabolites have emerged as key players in the pathogenesis of neuropsychiatric disorders. Recently, we demonstrated that animals susceptible to Single Prolonged Stress (SPS) have an overall pro-inflammatory gut microbiota and significantly lower cecal acetate levels than SPS-resilient rats, which correlated inversely with the anxiety index. Here, we investigated whether the microbial metabolite, acetate, could ameliorate SPS-triggered impairments. Male rats were randomly divided into unstressed controls or groups exposed to SPS. The groups received continued oral supplementation of either 150 mM of sodium acetate or 150 mM of sodium chloride-matched water. Two weeks after SPS, a battery of behavioral tests was performed, and the animals were euthanized the following day. While not affecting the unstressed controls, acetate supplementation reduced the impact of SPS on body weight gain and ameliorated SPS-induced anxiety-like behavior and the impairments in social interaction, but not depressive-like behavior. These changes were accompanied by several beneficial effects of acetate supplementation. Acetate alleviated the stress response by reducing urinary epinephrine levels, induced epigenetic modification by decreasing histone deacetylase (HDAC2) gene expression, inhibited neuroinflammation by reducing the density of Iba1+ cells and the gene expression of IL-1ß in the hippocampus, and increased serum β-hydroxybutyrate levels. The findings reveal a causal relationship between oral acetate treatment and mitigation of several SPS-induced behavioral impairments. Mechanistically, it impacted neuronal and metabolic pathways including changes in stress response, epigenetic modifications, neuroinflammation and showed novel link to ketone body production. The study demonstrates the preventive-therapeutic potential of acetate supplementation to alleviate adverse responses to traumatic stress.
Collapse
Affiliation(s)
- Arax Tanelian
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
| | - Bistra Nankova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
- Division of Newborn Medicine, Departments of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Furong Hu
- Division of Newborn Medicine, Departments of Pediatrics, New York Medical College, Valhalla, NY, USA
| | - Jordan D. Sahawneh
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
| | - Esther L. Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, USA
- Department of Psychiatry and Behavioral Science, New York Medical College, Valhalla, NY, USA
| |
Collapse
|
14
|
Alshehri A, Al-iedani O, Koussis N, Khormi I, Lea R, Lechner-Scott J, Ramadan S. Stability of longitudinal DTI metrics in MS with treatment of injectables, fingolimod and dimethyl fumarate. Neuroradiol J 2023; 36:388-396. [PMID: 36395524 PMCID: PMC10588600 DOI: 10.1177/19714009221140511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/20/2023] Open
Abstract
BACKGROUND AND PURPOSE Diffusion MRI (dMRI) is sensitive to microstructural changes in white matter of people with relapse-remitting multiple sclerosis (pw-RRMS) that lead to progressive disability. The role of diffusion in assessing the efficacy of different therapies requires more investigation. This study aimed to evaluate selected dMRI metrics in normal-appearing white matter and white matter-lesion in pw-RRMS and healthy controls longitudinally and compare the effect of therapies given. MATERIAL AND METHODS Structural and dMRI scans were acquired from 78 pw-RRMS (29 injectables, 36 fingolimod, 13 dimethyl fumarate) and 43 HCs at baseline and 2-years follow-up. Changes in dMRI metrics and correlation with clinical parameters were evaluated. RESULTS Differences were observed in most clinical parameters between pw-RRMS and HCs at both timepoints (p ≤ 0.01). No significant differences in average changes over time were observed for any dMRI metric between treatment groups in either tissue type. Diffusion metrics in NAWM and WML correlated negatively with most cognitive domains, while FA correlated positively at baseline but only for NAWM at follow-up (p ≤ 0.05). FA correlated negatively with disability in NAWM and WML over time, while MD and RD correlated positively only in NAWM. CONCLUSIONS This is the first DTI study comparing the effect of different treatments on dMRI parameters over time in a stable cohort of pw-RRMS. The results suggest that brain microstructural changes in a stable MS cohort are similar to HCs independent of the therapies used.
Collapse
Affiliation(s)
- Abdulaziz Alshehri
- School of Health Sciences, University of Newcastle College of Health, Medicine and Wellbeing, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Radiology, Imam Abdulrahman Bin Faisal University King Fahd University Hospital, Dammam, Saudi Arabia
| | - Oun Al-iedani
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Biomedical Sciences and Pharmacy, University of Newcastle College of Health, Medicine and Wellbeing, Callaghan, NSW, Australia
| | - Nikitas Koussis
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- School of Psychological Sciences, University of Newcastle College of Health, Medicine and Wellbeing, Callaghan, NSW, Australia
| | - Ibrahim Khormi
- School of Health Sciences, University of Newcastle College of Health, Medicine and Wellbeing, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- College of Applied Medical Sciences, University of Jeddah, Jeddah, Saudi Arabia
| | - Rodney Lea
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| | - Jeannette Lechner-Scott
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
- Department of Neurology, John Hunter Hospital, New Lambton Heights, NSW, Australia
- School of Medicine and Public Health, University of Newcastle College of Health, Medicine and Wellbeing, Callaghan, NSW, Australia
| | - Saadallah Ramadan
- School of Health Sciences, University of Newcastle College of Health, Medicine and Wellbeing, Callaghan, NSW, Australia
- Hunter Medical Research Institute, New Lambton Heights, NSW, Australia
| |
Collapse
|
15
|
Jamali E, Shapoori S, Farrokhi MR, Vakili S, Rostamzadeh D, Iravanpour F, Tavakoli Oliaee R, Jafarinia M. Effect of Disease-Modifying Therapies on COVID-19 Vaccination Efficacy in Multiple Sclerosis Patients: A Comprehensive Review. Viral Immunol 2023; 36:368-377. [PMID: 37276047 DOI: 10.1089/vim.2023.0035] [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] [Indexed: 06/07/2023] Open
Abstract
According to current knowledge, the etiopathogenesis of multiple sclerosis (MS) is complex, involving genetic background as well as several environmental factors that result in dysimmunity in the central nervous system (CNS). MS is an immune-mediated, inflammatory neurological disease affecting the CNS. As part of its attack on the axons of the CNS, MS witnesses varying degrees of myelin and axonal loss. A total of about 20 disease-modifying therapies (DMTs) are available today that, both in clinical trials and in real-world studies, reduce disease activity, such as relapses, magnetic resonance imaging lesions, and disability accumulation. Currently, the world is facing an outbreak of the new coronavirus disease 2019 (COVID-19), which originated in Wuhan, Hubei Province, China, in December 2019 and spread rapidly around the globe. Viral infections play an important role in triggering and maintaining neuroinflammation through direct and indirect mechanisms. There is an old association between MS and viral infections. In the context of MS-related chronic inflammatory damage within the CNS, there has been concern regarding COVID-19 worsening neurological damage. A high rate of disability and increased susceptibility to infection have made MS patients particularly vulnerable. In addition, DMTs have been a concern during the pandemic since many DMTs have immunosuppressive properties. In this article, we discuss the impact of DMTs on COVID-19 risks and the effect of DMTs on COVID-19 vaccination efficacy and outcome in MS patients.
Collapse
Affiliation(s)
- Elham Jamali
- Diagnostic Laboratory Sciences and Technology Research Center, School of Paramedical Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Peyvand Pathobiology and Genetic Laboratory, Shiraz, Iran
| | - Shima Shapoori
- Science Foundation Ireland (SFI), Center for Research in Medical Devices (CÚRAM), University of Galway, Galway, Ireland
| | - Majid Reza Farrokhi
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Neurosurgery, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sina Vakili
- Infertility Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Davoud Rostamzadeh
- Medicinal Plants Research Center, Yasuj University of Medical Sciences, Yasuj, Iran
- Department of Immunology, University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Farideh Iravanpour
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Razieh Tavakoli Oliaee
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Morteza Jafarinia
- Shiraz Neuroscience Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
16
|
Karaahmet B, Olschowka JA, O'Banion MK. Inconsistent Effects of Glatiramer Acetate Treatment in the 5xFAD Mouse Model of Alzheimer's Disease. Pharmaceutics 2023; 15:1809. [PMID: 37513996 PMCID: PMC10383120 DOI: 10.3390/pharmaceutics15071809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 06/17/2023] [Accepted: 06/20/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder that involves strong inflammatory components. Aberrant and prolonged inflammation in the CNS is thought to contribute to the development of the pathology. The use of single cytokine approaches to curb or leverage inflammatory mechanisms for disease modifying benefit has often resulted in conflicting data. Furthermore, these treatments were usually delivered locally into the CNS parenchyma, complicating translational efforts. To overcome these hurdles, we tested the use of glatiramer acetate (GA) in reducing amyloid beta (Aβ) plaque pathology in the 5xFAD model of AD. GA immunizations were begun at the ages of 2.5 months, 5.5 months, and 8.5 months, and GA was delivered weekly for 8 weeks. While previous data describe potential benefits of GA immunization in decreasing Aβ levels in murine models of AD, we found modest decreases in Aβ levels if given during the development of pathology but, surprisingly, found increased Aβ levels if GA was administered at later stages. The impact of GA treatment was only significant for female mice. Furthermore, we observed no changes between microglial uptake of plaque, CD11c immunopositivity of microglia, or levels of TMEM119 and P2Ry12 on microglia. Overall, these data warrant exercising caution when aiming to repurpose GA for AD.
Collapse
Affiliation(s)
- Berke Karaahmet
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - John A Olschowka
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - M Kerry O'Banion
- Department of Neuroscience, Del Monte Institute for Neuroscience, University of Rochester Medical Center, Rochester, NY 14642, USA
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
17
|
Gouttefangeas C, Klein R, Maia A. The good and the bad of T cell cross-reactivity: challenges and opportunities for novel therapeutics in autoimmunity and cancer. Front Immunol 2023; 14:1212546. [PMID: 37409132 PMCID: PMC10319254 DOI: 10.3389/fimmu.2023.1212546] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 05/24/2023] [Indexed: 07/07/2023] Open
Abstract
T cells are main actors of the immune system with an essential role in protection against pathogens and cancer. The molecular key event involved in this absolutely central task is the interaction of membrane-bound specific T cell receptors with peptide-MHC complexes which initiates T cell priming, activation and recall, and thus controls a range of downstream functions. While textbooks teach us that the repertoire of mature T cells is highly diverse, it is clear that this diversity cannot possibly cover all potential foreign peptides that might be encountered during life. TCR cross-reactivity, i.e. the ability of a single TCR to recognise different peptides, offers the best solution to this biological challenge. Reports have shown that indeed, TCR cross-reactivity is surprisingly high. Hence, the T cell dilemma is the following: be as specific as possible to target foreign danger and spare self, while being able to react to a large spectrum of body-threatening situations. This has major consequences for both autoimmune diseases and cancer, and significant implications for the development of T cell-based therapies. In this review, we will present essential experimental evidence of T cell cross-reactivity, implications for two opposite immune conditions, i.e. autoimmunity vs cancer, and how this can be differently exploited for immunotherapy approaches. Finally, we will discuss the tools available for predicting cross-reactivity and how improvements in this field might boost translational approaches.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ) partner site Tübingen, Tübingen, Germany
| | - Reinhild Klein
- Department of Hematology, Oncology, Clinical Immunology and Rheumatology, University Hospital Tübingen, Tübingen, Germany
| | - Ana Maia
- Nencki Institute of Experimental Biology of the Polish Academy of Sciences, Warsaw, Poland
- Cluster of Excellence iFIT (EXC2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tübingen, Tübingen, Germany
| |
Collapse
|
18
|
Rentsendorj A, Raedschelders K, Fuchs DT, Sheyn J, Vaibhav V, Porritt RA, Shi H, Dagvadorj J, de Freitas Germano J, Koronyo Y, Arditi M, Black KL, Gaire BP, Van Eyk JE, Koronyo-Hamaoui M. Osteopontin depletion in macrophages perturbs proteostasis via regulating UCHL1-UPS axis and mitochondria-mediated apoptosis. Front Immunol 2023; 14:1155935. [PMID: 37325640 PMCID: PMC10266348 DOI: 10.3389/fimmu.2023.1155935] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 05/19/2023] [Indexed: 06/17/2023] Open
Abstract
Introduction Osteopontin (OPN; also known as SPP1), an immunomodulatory cytokine highly expressed in bone marrow-derived macrophages (BMMΦ), is known to regulate diverse cellular and molecular immune responses. We previously revealed that glatiramer acetate (GA) stimulation of BMMΦ upregulates OPN expression, promoting an anti-inflammatory, pro-healing phenotype, whereas OPN inhibition triggers a pro-inflammatory phenotype. However, the precise role of OPN in macrophage activation state is unknown. Methods Here, we applied global proteome profiling via mass spectrometry (MS) analysis to gain a mechanistic understanding of OPN suppression versus induction in primary macrophage cultures. We analyzed protein networks and immune-related functional pathways in BMMΦ either with OPN knockout (OPNKO) or GA-mediated OPN induction compared with wild type (WT) macrophages. The most significant differentially expressed proteins (DEPs) were validated using immunocytochemistry, western blot, and immunoprecipitation assays. Results and discussion We identified 631 DEPs in OPNKO or GA-stimulated macrophages as compared to WT macrophages. The two topmost downregulated DEPs in OPNKO macrophages were ubiquitin C-terminal hydrolase L1 (UCHL1), a crucial component of the ubiquitin-proteasome system (UPS), and the anti-inflammatory Heme oxygenase 1 (HMOX-1), whereas GA stimulation upregulated their expression. We found that UCHL1, previously described as a neuron-specific protein, is expressed by BMMΦ and its regulation in macrophages was OPN-dependent. Moreover, UCHL1 interacted with OPN in a protein complex. The effects of GA activation on inducing UCHL1 and anti-inflammatory macrophage profiles were mediated by OPN. Functional pathway analyses revealed two inversely regulated pathways in OPN-deficient macrophages: activated oxidative stress and lysosome-mitochondria-mediated apoptosis (e.g., ROS, Lamp1-2, ATP-synthase subunits, cathepsins, and cytochrome C and B subunits) and inhibited translation and proteolytic pathways (e.g., 60S and 40S ribosomal subunits and UPS proteins). In agreement with the proteome-bioinformatics data, western blot and immunocytochemical analyses revealed that OPN deficiency perturbs protein homeostasis in macrophages-inhibiting translation and protein turnover and inducing apoptosis-whereas OPN induction by GA restores cellular proteostasis. Taken together, OPN is essential for macrophage homeostatic balance via the regulation of protein synthesis, UCHL1-UPS axis, and mitochondria-mediated apoptotic processes, indicating its potential application in immune-based therapies.
Collapse
Affiliation(s)
- Altan Rentsendorj
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Koen Raedschelders
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Dieu-Trang Fuchs
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Julia Sheyn
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Vineet Vaibhav
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Rebecca A. Porritt
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Haoshen Shi
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | | | | | - Yosef Koronyo
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pediatrics, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Keith L. Black
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Jennifer E. Van Eyk
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Neurology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
19
|
Valles SL, Singh SK, Campos-Campos J, Colmena C, Campo-Palacio I, Alvarez-Gamez K, Caballero O, Jorda A. Functions of Astrocytes under Normal Conditions and after a Brain Disease. Int J Mol Sci 2023; 24:ijms24098434. [PMID: 37176144 PMCID: PMC10179527 DOI: 10.3390/ijms24098434] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Revised: 04/26/2023] [Accepted: 05/05/2023] [Indexed: 05/15/2023] Open
Abstract
In the central nervous system (CNS) there are a greater number of glial cells than neurons (between five and ten times more). Furthermore, they have a greater number of functions (more than eight functions). Glia comprises different types of cells, those of neural origin (astrocytes, radial glia, and oligodendroglia) and differentiated blood monocytes (microglia). During ontogeny, neurons develop earlier (at fetal day 15 in the rat) and astrocytes develop later (at fetal day 21 in the rat), which could indicate their important and crucial role in the CNS. Analysis of the phylogeny reveals that reptiles have a lower number of astrocytes compared to neurons and in humans this is reversed, as there have a greater number of astrocytes compared to neurons. These data perhaps imply that astrocytes are important and special cells, involved in many vital functions, including memory, and learning processes. In addition, astrocytes are involved in different mechanisms that protect the CNS through the production of antioxidant and anti-inflammatory proteins and they clean the extracellular environment and help neurons to communicate correctly with each other. The production of inflammatory mediators is important to prevent changes in brain homeostasis. On the contrary, excessive, or continued production appears as a characteristic element in many diseases, such as Alzheimer's disease (AD), amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and in neurodevelopmental diseases, such as bipolar disorder, schizophrenia, and autism. Furthermore, different drugs and techniques have been developed to reverse oxidative stress and/or excess of inflammation that occurs in many CNS diseases, but much remains to be investigated. This review attempts to highlight the functional relevance of astrocytes in normal and neuropathological conditions by showing the molecular and cellular mechanisms of their role in the CNS.
Collapse
Affiliation(s)
- Soraya L Valles
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Sandeep Kumar Singh
- Indian Scientific Education and Technology Foundation, Lucknow 226002, India
| | - Juan Campos-Campos
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Carlos Colmena
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Ignacio Campo-Palacio
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Kenia Alvarez-Gamez
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
| | - Oscar Caballero
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| | - Adrian Jorda
- Department of Physiology, School of Medicine, University of Valencia, Blasco Ibañez 15, 46010 Valencia, Spain
- Faculty of Nursing and Podiatry, University of Valencia, 46010 Valencia, Spain
| |
Collapse
|
20
|
Kihara Y, Chun J. Molecular and neuroimmune pharmacology of S1P receptor modulators and other disease-modifying therapies for multiple sclerosis. Pharmacol Ther 2023; 246:108432. [PMID: 37149155 DOI: 10.1016/j.pharmthera.2023.108432] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 04/25/2023] [Accepted: 05/02/2023] [Indexed: 05/08/2023]
Abstract
Multiple sclerosis (MS) is a neurological, immune-mediated demyelinating disease that affects people in the prime of life. Environmental, infectious, and genetic factors have been implicated in its etiology, although a definitive cause has yet to be determined. Nevertheless, multiple disease-modifying therapies (DMTs: including interferons, glatiramer acetate, fumarates, cladribine, teriflunomide, fingolimod, siponimod, ozanimod, ponesimod, and monoclonal antibodies targeting ITGA4, CD20, and CD52) have been developed and approved for the treatment of MS. All the DMTs approved to date target immunomodulation as their mechanism of action (MOA); however, the direct effects of some DMTs on the central nervous system (CNS), particularly sphingosine 1-phosphate (S1P) receptor (S1PR) modulators, implicate a parallel MOA that may also reduce neurodegenerative sequelae. This review summarizes the currently approved DMTs for the treatment of MS and provides details and recent advances in the molecular pharmacology, immunopharmacology, and neuropharmacology of S1PR modulators, with a special focus on the CNS-oriented, astrocyte-centric MOA of fingolimod.
Collapse
Affiliation(s)
- Yasuyuki Kihara
- Sanford Burnham Prebys Medical Discovery Institute, United States of America.
| | - Jerold Chun
- Sanford Burnham Prebys Medical Discovery Institute, United States of America
| |
Collapse
|
21
|
Monroy GR, Murguiondo Pérez R, Weintraub Ben Zión E, Vidal Alcántar-Garibay O, Loza-López EC, Tejerina Marion E, Blancarte Hernández E, Navarro-Torres L, Ibarra A. Immunization with Neural-Derived Peptides in Neurodegenerative Diseases: A Narrative Review. Biomedicines 2023; 11:biomedicines11030919. [PMID: 36979898 PMCID: PMC10046177 DOI: 10.3390/biomedicines11030919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 02/24/2023] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are a major health problem worldwide. Statistics suggest that in America in 2030 there will be more than 12 million people suffering from a neurodegenerative pathology. Furthermore, the increase in life expectancy enhances the importance of finding new and better therapies for these pathologies. NDDs could be classified into chronic or acute, depending on the time required for the development of clinical symptoms and brain degeneration. Nevertheless, both chronic and acute stages share a common immune and inflammatory pathway in their pathophysiology. Immunization with neural-derived peptides (INDP) is a novel therapy that has been studied during the last decade. By inoculating neural-derived peptides obtained from the central nervous system (CNS), this therapy aims to boost protective autoimmunity, an autoreactive response that leads to a protective phenotype that produces a healing environment and neuroregeneration instead of causing damage. INDP has shown promising findings in studies performed either in vitro, in vivo or even in some pre-clinical trials of different NDDs, standing as a potentially beneficial therapy. In this review, we will describe some of the studies in which the effect of INDP strategies have been explored in different (chronic and acute) neurodegenerative diseases.
Collapse
Affiliation(s)
- Germán Rivera Monroy
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Renata Murguiondo Pérez
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Efraín Weintraub Ben Zión
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Oscar Vidal Alcántar-Garibay
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Ericka Cristina Loza-López
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Emilio Tejerina Marion
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Enrique Blancarte Hernández
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Lisset Navarro-Torres
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
| | - Antonio Ibarra
- Centro de Investigación en Ciencias de la Salud (CICSA), FCS, Universidad Anáhuac México, Huixquilucan 52786, Mexico
- Neuroimmunology Department, Proyecto CAMINA A.C., Ciudad de México 14370, Mexico
- Correspondence:
| |
Collapse
|
22
|
Koronyo-Hamaoui M, Gaire BP, Frautschy SA, Alvarez JI. Editorial: Role of Inflammation in Neurodegenerative Diseases. Front Immunol 2022; 13:958487. [PMID: 35799792 PMCID: PMC9253757 DOI: 10.3389/fimmu.2022.958487] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 06/09/2022] [Indexed: 12/30/2022] Open
Affiliation(s)
- Maya Koronyo-Hamaoui
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Division of Applied Cell Biology and Physiology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- *Correspondence: Maya Koronyo-Hamaoui,
| | - Bhakta Prasad Gaire
- Department of Neurosurgery, Maxine Dunitz Neurosurgical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Sally Ann Frautschy
- Geriatric Research Education and Clinical Center, Veterans Greater Los Angeles HealthCare System, Los Angeles, CA, United States
- Department of Neurology, University of California, Los Angeles, Los Angeles, CA, United States
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Jorge Ivan Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|