1
|
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
|
2
|
Godínez-Chaparro B, Rodríguez-Ramos MC, Martínez-Lorenzana MG, González-Morales E, Pérez-Ruíz KP, Espinosa de Los Monteros-Zuñiga A, Mendoza-Pérez F, Condes-Lara M. Pramipexole decreases allodynia and hyperalgesia via NF-κB in astrocytes in rats with Parkinson's disease. Pharmacol Biochem Behav 2025; 247:173945. [PMID: 39675389 DOI: 10.1016/j.pbb.2024.173945] [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: 04/08/2024] [Revised: 10/30/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024]
Abstract
Pain is one of the principal non-motor symptoms of Parkinson's disease (PD), negatively impacting the patient's quality of life. This study aimed to demonstrate whether an effective dose of pramipexole (PPX) can modulate the NF-κB/p-p65 activation in glial cells (astrocytes and microglia) and diminish the hypersensitivity (allodynia and hyperalgesia) in male Wistar rats with PD. For this, 2 μl of 6-hydroxydopamine (6-OHDA, 8 μg/μL/0.2 μl/min) was administered unilaterally in the Substantia Nigra of the Pars Compacta (SNpc) to establish a PD model rat. Motor behavioral tests were used to validate the PD model, and von Frey filaments were used to evaluate allodynia and hyperalgesia. Immunohistochemical and immunofluorescence were used to analyze the level of tyrosine hydroxylase in SNpc and striatum as well as the expression of GFAP, Iba-1, NF-κB/p-65 in the L4-L6 spinal cord dorsal horn. Unilateral 6-OHDA-lesion reduces motor capacity and produces long-term allodynia and hyperalgesia in both hind paws. L4-L6 spinal cord dorsal horn astrocytes and microglia were active in these 6-OHDA-lesioned rats. Moreover, PPX (1 and 3 mg/Kg, i.p./10 days, n = 10 per group) inhibited the bilateral mechanical hypersensitivity, and PPX (3 mg/Kg/i.p./10 days) reduced 6-OHDA-induced astrocyte and microglia activation, as well as reduced NF-κB/p-p65 expression only in astrocytes of dorsal horn spinal cord in the L5-L6. These findings suggest that PPX could alleviate pain by decreasing the activation of microglia and astrocytes through the NF-κB/p-p65 pathway in the dorsal horn spinal cord. Therefore, PPX could be considered an optional tool for improving pain hypersensitivity in PD patients.
Collapse
Affiliation(s)
- Beatriz Godínez-Chaparro
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico.
| | - Maria Cristina Rodríguez-Ramos
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - María Guadalupe Martínez-Lorenzana
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Estefanía González-Morales
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - Karen Pamela Pérez-Ruíz
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - Antonio Espinosa de Los Monteros-Zuñiga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| | - Felipe Mendoza-Pérez
- Departamento de Sistemas Biológicos, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Xochimilco, Calzada del Hueso 1100, Colonia Villa Quietud, 04960, Mexico City, Mexico
| | - Miguel Condes-Lara
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus Juriquilla, Boulevard Juriquilla, No. 3001, C.P. 76230, Querétaro, Mexico
| |
Collapse
|
3
|
Raffaele S, Clausen BH, Mannella FC, Wirenfeldt M, Marangon D, Tidgen SB, Corradini S, Madsen K, Lecca D, Abbracchio MP, Lambertsen KL, Fumagalli M. Characterisation of GPR17-expressing oligodendrocyte precursors in human ischaemic lesions and correlation with reactive glial responses. J Pathol 2025; 265:226-243. [PMID: 39703181 PMCID: PMC11717493 DOI: 10.1002/path.6381] [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: 06/03/2024] [Revised: 10/14/2024] [Accepted: 11/20/2024] [Indexed: 12/21/2024]
Abstract
White matter damage and subsequent demyelination significantly contribute to long-term functional impairment after ischaemic stroke. Identifying novel pharmacological targets to restore myelin integrity by promoting the maturation of oligodendrocyte precursor cells (OPCs) into new myelinating oligodendrocytes may open new perspectives for ischaemic stroke treatment. In this respect, previous studies highlighted the role of the G protein-coupled membrane receptor 17 (GPR17) as a key regulator of OPC differentiation in experimental models of brain injury, including ischaemic stroke. To determine the translational value of GPR17 as a possible target in the context of human disease, we exploited immunohistochemistry to characterise the distribution of GPR17-expressing cells in brain tissue samples from ischaemic stroke cases and correlated it with the reactive state of neighbouring glial cells. The results showed that GPR17 specifically decorates a subpopulation of differentiation-committed OPCs, labelled by the peculiar marker breast carcinoma-amplified sequence 1 (BCAS1), that accumulates in the peri-infarct region in the later stages after the ischaemic event. Interestingly, the response of GPR17-expressing cells appears to be paralleled by the switch of reactive microglia/macrophages from a phagocytic to a dystrophic phenotype and by astrocytic scar formation. A negative correlation was found between GPR17-expressing OPCs and reactive microglia/macrophages and astrocytes surrounding chronic ischaemic lesions in female subjects, while the same relationship was less pronounced in males. These results were reinforced by bioinformatic analysis of a publicly available transcriptomic dataset, which implicated a possible role of inflammation and defective neuron-to-OPC communication in remyelination failure after ischaemic damage. Hence, these data strengthen the relevance of GPR17-based remyelinating therapies for the treatment of ischaemic stroke. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Stefano Raffaele
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| | - Bettina Hjelm Clausen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
| | - Francesca Carolina Mannella
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| | - Martin Wirenfeldt
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
- Department of PathologySouth Denmark University HospitalOdenseDenmark
| | - Davide Marangon
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | - Sarah Boe Tidgen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Silvia Corradini
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Kirsten Madsen
- Department of PathologySouth Denmark University HospitalOdenseDenmark
- Department of Cardiovascular and Renal Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Davide Lecca
- Department of Pharmaceutical SciencesUniversità degli Studi di MilanoMilanItaly
| | | | - Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Department of Clinical Research, Brain Research – Inter Disciplinary Guided Excellence (BRIDGE)University of Southern DenmarkOdenseDenmark
- Odense Patient data Explorative Network (OPEN), Department of Clinical Research, Odense University HospitalUniversity of Southern DenmarkOdenseDenmark
- Department of NeurologyOdense University HospitalOdenseDenmark
| | - Marta Fumagalli
- Department of Pharmacological and Biomolecular Sciences ‘Rodolfo Paoletti’Università degli Studi di MilanoMilanItaly
| |
Collapse
|
4
|
Bobotis BC, Khakpour M, Braniff O, de Andrade EG, Gargus M, Allen M, Carrier M, Baillargeon J, Rangachari M, Tremblay MÈ. Sex chromosomes and sex hormones differently shape microglial properties during normal physiological conditions in the adult mouse hippocampus. J Neuroinflammation 2025; 22:18. [PMID: 39856696 PMCID: PMC11762133 DOI: 10.1186/s12974-025-03341-6] [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: 10/16/2024] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
The brain presents various structural and functional sex differences, for which multiple factors are attributed: genetic, epigenetic, metabolic, and hormonal. While biological sex is determined by both sex chromosomes and sex hormones, little is known about how these two factors interact to establish this dimorphism. Sex differences in the brain also affect its resident immune cells, microglia, which actively survey the brain parenchyma and interact with sex hormones throughout life. However, microglial differences in density and distribution, morphology and ultrastructural patterns in physiological conditions during adulthood are largely unknown. Here, we investigated these aforementioned properties of microglia using the Four Core Genotypes (FCG) model, which allows for an independent assessment of gonadal hormones and sex chromosomal effects in four conditions: FCG XX and Tg XY- (both ovaries); Tg XXSry and Tg XYSry (both testes). We also compared the FCG results with XX and XY wild-type (WT) mice. In adult mice, we focused our investigation on the ventral hippocampus across different layers: CA1 stratum radiatum (Rad) and CA1 stratum lacunosum-moleculare (LMol), as well as the dentate gyrus polymorphic layer (PoDG). Double immunostaining for Iba1 and TMEM119 revealed that microglial density is influenced by both sex chromosomes and sex hormones. We show in the Rad and LMol that microglia are denser in FCG XX compared to Tg XYSry mice, however, microglia were densest in WT XX mice. In the PoDG, ovarian animals had increased microglial density compared to testes animals. Additionally, microglial morphology was modulated by a complex interaction between hormones and chromosomes, affecting both their cellular soma and arborization across the hippocampal layers. Moreover, ultrastructural analysis showed that microglia in WT animals make overall more contacts with pre- and post-synaptic elements than in FCG animals. Lastly, microglial markers of cellular stress, including mitochondrion elongation, and dilation of the endoplasmic reticulum and Golgi apparatus, were mostly chromosomally driven. Overall, we characterized different aspects of microglial properties during normal physiological conditions that were found to be shaped by sex chromosomes and sex hormones, shading more light onto how sex differences affect the brain immunity at steady-state.
Collapse
Affiliation(s)
- Bianca Caroline Bobotis
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Mohammadparsa Khakpour
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | | | - Makenna Gargus
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Micah Allen
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Joanie Baillargeon
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Manu Rangachari
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada.
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada.
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
5
|
Chen XF, Kroke B, Ni J, Munoz C, Appleman M, Jacobs B, Tran T, Nguyen KV, Qiu C, Stonestreet BS, Marshall J. Novel peptidomimetic compounds attenuate hypoxic-ischemic brain injury in neonatal rats. Exp Neurol 2025; 386:115151. [PMID: 39832663 DOI: 10.1016/j.expneurol.2025.115151] [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: 10/21/2024] [Revised: 01/04/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Hypoxic-ischemic (HI) brain injury is a common neurological problem in neonates. The postsynaptic density protein-95 (PSD-95) is an excitatory synaptic scaffolding protein that regulates synaptic function, and represents a potential therapeutic target to attenuate HI brain injury. Syn3 and d-Syn3 are novel high affinity cyclic peptides that bind the PDZ3 domain of PSD-95. We investigated the neuroprotective efficacy of Syn3 and d-Syn3 after exposure to HI in neonatal rodents. Postnatal (P) day-7 rats were treated with Syn3 and d-Syn3 at zero, 24, and 48-h after carotid artery ligation and 90-min of 8 % oxygen. Hemispheric volume atrophy and Iba-1 positive microglia were quantified by cresyl violet and immunohistochemical staining. Treatment with Syn3 and d-Syn3 reduced tissue volume loss by 47.0 % and 41.0 % in the male plus female, and by 42.1 % and 65.0 % in the male groups, respectively. Syn3 reduced tissue loss by 52.3 % in females. D-Syn3 reduced Iba-1 positive microglia/DAPI ratios in the pooled group, males, and females. Syn3 effects were observed in the pooled group and females. Changes in Iba-1 positive microglia/DAPI cellular ratios correlated directly with reduced hemispheric volume loss, suggesting that Syn3 and d-Syn3 provide neuroprotection in part by their effects on Iba-1 positive microglia. The pathogenic cis phosphorylated Thr231 in Tau (cis P-tau) is a marker of neuronal injury. Cis P-tau was induced in cortical cells of the placebo-treated pooled group, males and females after HI, and reduced by treatment with d-Syn3. Therefore, treatment with these peptidomimetic agents exert neuroprotective effects after exposure of neonatal subjects to HI related brain injury.
Collapse
Affiliation(s)
- Xiaodi F Chen
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA.
| | - Brynn Kroke
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Jun Ni
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christian Munoz
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Mark Appleman
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Bryce Jacobs
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA
| | - Tuong Tran
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Kevin V Nguyen
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - Chenxi Qiu
- Department of Neurology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Barbara S Stonestreet
- Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA; Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA; Emerita, Department of Pediatrics, Women & Infants Hospital of RI, The Alpert Medical School of Brown University, Providence, RI, USA
| | - John Marshall
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI, USA.
| |
Collapse
|
6
|
Feichtenbiner AB, Sytsma K, O'Boyle RP, Mittenzwei R, Maioli H, Scherpelz KP, Child DD, Li N, Ariza Torres J, Keene L, Kirkland A, Howard K, Latimer C, Keene CD, Ransom C, Nolan AL. Satellite microglia: marker of traumatic brain injury and regulator of neuronal excitability. J Neuroinflammation 2025; 22:9. [PMID: 39819341 PMCID: PMC11740464 DOI: 10.1186/s12974-024-03328-9] [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: 10/18/2024] [Accepted: 12/24/2024] [Indexed: 01/19/2025] Open
Abstract
Traumatic brain injury is a leading cause of chronic neurologic disability and a risk factor for development of neurodegenerative disease. However, little is known regarding the pathophysiology of human traumatic brain injury, especially in the window after acute injury and the later life development of progressive neurodegenerative disease. Given the proposed mechanisms of toxic protein production and neuroinflammation as possible initiators or contributors to progressive pathology, we examined phosphorylated tau accumulation, microgliosis and astrogliosis using immunostaining in the orbitofrontal cortex, a region often vulnerable across traumatic brain injury exposures, in an age and sex-matched cohort of community traumatic brain injury including both mild and severe cases in midlife. We found that microglial response is most prominent after chronic traumatic brain injury, and interactions with neurons in the form of satellite microglia are increased, even after mild traumatic brain injury. Taking our investigation into a mouse model, we identified that these satellite microglia suppress neuronal excitability in control conditions but lose this ability with chronic traumatic brain injury. At the same time, network hyperexcitability is present in both mouse and human orbitofrontal cortex. Our findings support a role for loss of homeostatic control by satellite microglia in the maladaptive circuit changes that occur after traumatic brain injury.
Collapse
Affiliation(s)
- Alicia B Feichtenbiner
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Karinn Sytsma
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Ryan P O'Boyle
- Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
| | - Rhonda Mittenzwei
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
- King County Office of the Medical Examiner, Seattle, WA, 98104, USA
| | - Heather Maioli
- Office of Chief Medical Examiner of the City of New York, New York, NY, 10016, USA
| | - Kathryn P Scherpelz
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Daniel D Child
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Ning Li
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA
| | | | - Lisa Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Amanda Kirkland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Kimberly Howard
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Caitlin Latimer
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - C Dirk Keene
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA
| | - Christopher Ransom
- Department of Neurology, University of Washington, Seattle, WA, 98195, USA
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA
| | - Amber L Nolan
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98104, USA.
- Puget Sound Veterans Affairs Seattle Medical Center, Seattle, WA, 98108, USA.
| |
Collapse
|
7
|
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
|
8
|
Robb JL, Boisjoly F, Machuca-Parra AI, Coursan A, Manceau R, Majeur D, Rodaros D, Bouyakdan K, Greffard K, Bilodeau JF, Forest A, Daneault C, Ruiz M, Laurent C, Arbour N, Layé S, Fioramonti X, Madore C, Fulton S, Alquier T. Blockage of ATGL-mediated breakdown of lipid droplets in microglia alleviates neuroinflammatory and behavioural responses to lipopolysaccharides. Brain Behav Immun 2025; 123:315-333. [PMID: 39326768 DOI: 10.1016/j.bbi.2024.09.027] [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: 02/05/2024] [Revised: 08/21/2024] [Accepted: 09/21/2024] [Indexed: 09/28/2024] Open
Abstract
Lipid droplets (LD) are triglyceride storing organelles that have emerged as an important component of cellular inflammatory responses. LD lipolysis via adipose triglyceride lipase (ATGL), the enzyme that catalyses the rate-limiting step of triglyceride lipolysis, regulates inflammation in peripheral immune and non-immune cells. ATGL elicits both pro- and anti-inflammatory responses in the periphery in a cell-type dependent manner. The present study determined the impact of ATGL inhibition and microglia-specific ATGL genetic loss-of-function on acute inflammatory and behavioural responses to pro-inflammatory insult. First, we evaluated the impact of lipolysis inhibition on lipopolysaccharide (LPS)-induced expression and secretion of cytokines and phagocytosis in mouse primary microglia cultures. Lipase inhibitors (ORlistat and ATGListatin) and LPS led to LD accumulation in microglia. Pan-lipase inhibition with ORlistat alleviated LPS-induced expression of IL-1β and IL-6. Specific inhibition of ATGL had a similar action on CCL2, IL-1β and IL-6 expression in both neonatal and adult microglia cultures. CCL2 and IL-6 secretion were also reduced by ATGListatin or knockdown of ATGL. ATGListatin increased phagocytosis in neonatal cultures independently from LPS treatment. Second, targeted and untargeted lipid profiling revealed that ATGListatin reduced LPS-induced generation of pro-inflammatory prostanoids and modulated ceramide species in neonatal microglia. Finally, the role of microglial ATGL in neuroinflammation was assessed using a novel microglia-specific and inducible ATGL knockout mouse model. Loss of microglial ATGL in adult male mice dampened LPS-induced expression of IL-6 and IL-1β and microglial density. LPS-induced sickness- and anxiety-like behaviours were also reduced in male mice with loss of ATGL in microglia. Together, our results demonstrate potent anti-inflammatory effects produced by pharmacological or genetic inhibition of ATGL-mediated triglyceride lipolysis and thereby propose that supressing microglial LD lipolysis has beneficial actions in acute neuroinflammatory conditions.
Collapse
Affiliation(s)
- Josephine Louise Robb
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Frédérick Boisjoly
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Arturo Israel Machuca-Parra
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Adeline Coursan
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Romane Manceau
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Danie Majeur
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Demetra Rodaros
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Khalil Bouyakdan
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Karine Greffard
- Axe Endocrinologie et Néphrologie, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Jean-François Bilodeau
- Axe Endocrinologie et Néphrologie, CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada; Département de Médecine, Faculté de Médecine, Université Laval, Québec, QC, G1K 7P4, Canada
| | - Anik Forest
- Institut de Cardiologie de Montréal, Plateforme de métabolomique, Montréal, QC H1T1C8, Canada
| | - Caroline Daneault
- Institut de Cardiologie de Montréal, Plateforme de métabolomique, Montréal, QC H1T1C8, Canada
| | - Matthieu Ruiz
- Département de Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada; Institut de Cardiologie de Montréal, Plateforme de métabolomique, Montréal, QC H1T1C8, Canada
| | - Cyril Laurent
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Nathalie Arbour
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Neurosciences, Université de Montréal, Montréal, QC H3T 1J4, Canada
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Xavier Fioramonti
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Charlotte Madore
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Stephanie Fulton
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Nutrition, Université de Montréal, Montréal, QC H3T 1J4, Canada; Food4BrainHealth France-Canada International Research Network, Bordeaux, France
| | - Thierry Alquier
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Université de Montréal, Montréal, QC H3T 1J4, Canada; Département de Médecine, Université de Montréal, Montréal, QC H3T 1J4, Canada; Food4BrainHealth France-Canada International Research Network, Bordeaux, France.
| |
Collapse
|
9
|
King DP, Abdalaziz M, Majewska AK, Cameron JL, Fudge JL. Microglia Morphology in the Developing Primate Amygdala and Effects of Early Life Stress. eNeuro 2025; 12:ENEURO.0466-24.2024. [PMID: 39753372 PMCID: PMC11735683 DOI: 10.1523/eneuro.0466-24.2024] [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: 10/28/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/18/2025] Open
Abstract
A unique pool of immature glutamatergic neurons in the primate amygdala, known as the paralaminar nucleus (PL), are maturing between infancy and adolescence. The PL is a potential substrate for the steep growth curve of amygdala volume during this developmental period. A microglial component is also embedded among the PL neurons and likely supports local neuronal maturation and emerging synaptogenesis. Microglia may alter neuronal growth following environmental perturbations such as stress. Using multiple measures in rhesus macaques, we found that microglia in the infant primate PL had relatively large somas and a small arbor size. In contrast, microglia in the adolescent PL had a smaller soma and a larger dendritic arbor. We then examined microglial morphology in the PL after a novel maternal separation protocol, to examine the effects of early life stress. After maternal separation, the microglia had increased soma size, arbor size, and complexity. Surprisingly, strong effects were seen not only in the infant PL, but also in the adolescent PL from subjects who had experienced the separation many years earlier. We conclude that under normal maternal-rearing conditions, PL microglia morphology tracks PL neuronal growth, progressing to a more "mature" phenotype by adolescence. Maternal separation has long-lasting effects on microglia, altering their normal developmental trajectory, and resulting in a "hyper-ramified" phenotype that persists for years. We speculate that these changes have consequences for neuronal development in young primates.
Collapse
Affiliation(s)
- Dennisha P King
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Miral Abdalaziz
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Ania K Majewska
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
| | - Judy L Cameron
- Department of Psychiatry, University of Pittsburgh, Pittsburgh, Pennsylvania 15213
| | - Julie L Fudge
- Department of Neuroscience, University of Rochester Medical Center, Rochester, New York 14642
- Department of Psychiatry, University of Rochester Medical Center, Rochester, New York 14642
| |
Collapse
|
10
|
Bortolin V, Mansuroglu Z, Conquet L, Calcagno G, Lambert F, Marin-Obando JP, Segrt H, Savino M, Menidjel R, Souès S, Buée L, Niedergang F, Galas MC, Montagutelli X, Bonnefoy E. Protein kinase R induced by type I interferons is a main regulator of reactive microglia in Zika virus infection. Glia 2025; 73:80-104. [PMID: 39359232 DOI: 10.1002/glia.24619] [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: 04/26/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024]
Abstract
Microglial cells are the phagocytic cells of the brain that under physiological conditions participate in brain homeostasis and surveillance. Under pathogenic states, microglia undergoes strong morphological and transcriptional changes potentially leading to sustained neuroinflammation, brain damage, and cognitive disorders. Postnatal and adult Zika virus (ZIKV) brain infection is characterized by the induction of reactive microglia associated with brain inflammation, synapse loss and neuropathogenesis. Contrary to neurons, microglial cells are not infected by ZIKV thus raising the question of the mechanism governing ZIKV-induced microglia's reactivity. In this work, we have questioned the role of exogenous, neuronal type I interferons (IFNs-I) in regulating ZIKV-induced microglia's reactivity. Primary cultured microglial cells were either treated with conditioned media from ZIKV-infected mature neurons or co-cultured with ZIKV-infected neurons. Using either an antibody directed against the IFNAR receptor that neutralizes the IFNs-I response or Ifnar-/-microglial cells, we demonstrate that IFNs-I produced by ZIKV-infected neurons are the main regulators of the phagocytic capacity and the pro-inflammatory gene expression profile of reactive, non-infected microglial cells. We identify protein kinase R (PKR), whose expression is activated by IFNs-I, as a major regulator of the phagocytic capacity, pro-inflammatory response, and morphological changes of microglia induced by IFNs-I while up-regulating STAT1 phosphorylation and IRF1 expression. Results obtained herein in vitro with primary cultured cells and in vivo in ZIKV-infected adult immunocompetent mice, unravel a role for IFNs-I and PKR in directly regulating microglia's reactivity that could be at work in other infectious and non-infectious brain pathologies.
Collapse
Affiliation(s)
| | - Zeyni Mansuroglu
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Laurine Conquet
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Gaetano Calcagno
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Fanny Lambert
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | | | - Helena Segrt
- Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, University of Lille, Lille, France
| | - Mary Savino
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Reyene Menidjel
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Sylvie Souès
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| | - Luc Buée
- Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, University of Lille, Lille, France
| | | | - Marie-Christine Galas
- Inserm, CHU Lille, CNRS, LilNCog-Lille Neuroscience & Cognition, University of Lille, Lille, France
| | - Xavier Montagutelli
- Institut Pasteur, Université Paris Cité, Mouse Genetics Laboratory, Paris, France
| | - Eliette Bonnefoy
- CNRS, Inserm, Institut Cochin, Université Paris Cité, Paris, France
| |
Collapse
|
11
|
Kendall G, Underwood C, Parr‐Brownlie L. A Novel Rat Model for Inflammatory Gut-Brain Interactions in Parkinson's Disease. Eur J Neurosci 2025; 61:e16667. [PMID: 39844519 PMCID: PMC11754928 DOI: 10.1111/ejn.16667] [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: 07/03/2024] [Revised: 12/16/2024] [Accepted: 12/22/2024] [Indexed: 01/24/2025]
Abstract
Gut inflammation is a salient prodromal feature of Parkinson's disease (PD) implicated in pathologic processes leading to nigrostriatal dopaminergic degeneration. However, existing rodent models of PD are suboptimal for investigating the interaction between gut inflammation and neuropathology. This study aimed to develop a rat model of PD in which gut inflammation exacerbated PD symptoms induced by a parkinsonian lesion. This was achieved by combining the 6-hydroxydopamine (6-OHDA) rat model for PD and the dextran sodium sulfate (DSS) rat model for colitis. The model was characterised using behavioural tests, including reaching, step, gait, open-field and cylinder tests, plus stereological quantification of substantia nigra (SN) DA neurodegeneration, and histological analysis of SN microglial activation and distal colon morphology. The combination of 6-OHDA and DSS resulted in greater stool softening and bleeding, shorter colons and greater distal colon histological damage, when compared with the 6-OHDA model. Additionally, 6-OHDA and DSS rats displayed similar DA neurodegeneration, yet less SN microglial activation, when compared to 6-OHDA rats that did not receive DSS. Finally, DSS + 6-OHDA rats exhibited impaired forelimb motor function compared with 6-OHDA rats, with decreased performance in reaching and step tests. In conclusion, DSS administration exacerbated forelimb motor dysfunction in 6-OHDA rats. Behavioural changes in DSS + 6-OHDA rats were associated with lower levels of microglial activation and similar levels of dopamine depletion compared with 6-OHDA-only rats. These results support that the DSS + 6-OHDA rat model is a promising PD animal model to investigate deleterious gut-brain interactions in PD.
Collapse
Affiliation(s)
- Grace E. Kendall
- Department of Anatomy, Brain Health Research CentreUniversity of OtagoDunedinNew Zealand
| | - Conor F. Underwood
- Department of Anatomy, Brain Health Research CentreUniversity of OtagoDunedinNew Zealand
| | | |
Collapse
|
12
|
Gonul CP, Kiser C, Yaka EC, Oz D, Hunerli D, Yerlikaya D, Olcum M, Keskinoglu P, Yener G, Genc S. Microglia-like cells from patient monocytes demonstrate increased phagocytic activity in probable Alzheimer's disease. Mol Cell Neurosci 2024:103990. [PMID: 39732446 DOI: 10.1016/j.mcn.2024.103990] [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: 10/25/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder that is characterized by the accumulation of amyloid plaques, phosphorylated tau tangles and microglia toxicity, resulting in neuronal death and cognitive decline. Since microglia are recognized as one of the key players in the disease, it is crucial to understand how microglia operate in disease conditions and incorporate them into models. The studies on human microglia functions are thought to reflect the post-symptomatic stage of the disease. Recently developed methods involve induced microglia-like cells (iMGs) generated from patients' blood monocytes or induced pluripotent stem cells (iPSCs) as an alternative to studying the microglia cells in vitro. In this research, we aimed to investigate the phenotype and inflammatory responses of iMGs from AD patients. Monocytes derived from blood using density gradient centrifugation were differentiated into iMGs using a cytokine cocktail, including granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-34 (IL-34). After differentiation, cells were assessed by morphological analysis and a microglia surface marker, TMEM119. We used stimulants, lipopolysaccharide (LPS) and beta-amyloid, to examine iMGs' functions. Results showed that iMGs derived from AD patients exhibited increased secretion of pro-inflammatory cytokines upon LPS stimulation. Furthermore, their phagocytic ability was also heightened in stimulated and unstimulated conditions, with cells derived from patients showing increased phagocytic activity compared to healthy controls. Overall, these findings suggest that iMGs derived from patients using the direct conversion method possess characteristics of human microglia, making them an easy and promising model for studying microglia function in AD.
Collapse
Affiliation(s)
- Ceren Perihan Gonul
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Cagla Kiser
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye
| | - Emis Cansu Yaka
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye; Department of Neurology, İzmir City Hospital, Izmir, Türkiye
| | - Didem Oz
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye; Department of Neurology, Dokuz Eylul University Hospital, Izmir, Türkiye; Global Brain Health Institute, University of California, San Francisco, USA
| | - Duygu Hunerli
- Department of Biostatistics and Medical Informatics, Basic Medical Sciences, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Deniz Yerlikaya
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye
| | - Melis Olcum
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye
| | - Pembe Keskinoglu
- Department of Biostatistics and Medical Informatics, Basic Medical Sciences, Faculty of Medicine, Dokuz Eylul University, Izmir, Türkiye
| | - Gorsev Yener
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Department of Neurology, Dokuz Eylul University Hospital, Izmir, Türkiye
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Türkiye; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Türkiye; Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University, Izmir, Türkiye.
| |
Collapse
|
13
|
Rao B, Liu X, Xiao J, Wu X, He F, Yang Q, Zhao W, Lin X, Zhang J. Microglia heterogeneity during neuroinflammation and neurodegeneration in the mouse retina. Brain Struct Funct 2024; 230:19. [PMID: 39720969 DOI: 10.1007/s00429-024-02875-4] [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: 06/13/2024] [Accepted: 10/15/2024] [Indexed: 12/26/2024]
Abstract
Microglia play important roles in maintaining homeostasis and immunoreactive defense in the central nervous system including retina. To accomplish such a wide range of functions, microglia are highly heterogeneous. Dark microglia (DM) were recently identified by electron microscopy (EM). However, the specific correlation between microglial morphological phenotypes, including DM, and physiological or pathological conditions remains poorly understood. We established acute and chronic neuroinflammatory models by Lipopolysaccharide (LPS) and light-induced photoreceptor neurodegeneration model to explore these questions in the mouse retina. Immunofluorescence and EM were used to detect microglia in these models. Our light microscopy (LM) results reveal that the withdrawal phenotype is predominant in acute neuroinflammation models, both in vitro and in vivo, while the dystrophic microglia are the major phenotype in chronic neuroinflammation and neurodegeneration models in vivo. Ultrastructurally, acute models exhibit high electron dense processes, but not somas, while chronic models show high electron dense somas and processes. Given the consistency between LM and EM, we propose that DM-like somas and processes likely indicate a dystrophic population. It's important to note, however, that DM may not represent a single specific microglia phenotype, but rather a dynamic transformation of gradually activated microglia. Finally, we provide evidence for the presence of DM in mouse retinas in the neuroinflammatory model and the neurodegenerative model. This research provides valuable insights into investigating microglia phenotypes through both LM and EM.
Collapse
Affiliation(s)
- Bilin Rao
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaoqing Liu
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiayi Xiao
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xiaotian Wu
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Fang He
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Qingwen Yang
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Wenna Zhao
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Xin Lin
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China
| | - Jun Zhang
- State Key Laboratory of Ophthalmology, Optometry and Visual Science, Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
- Laboratory of Retinal Physiology and Disease, School of Ophthalmology and Optometry and Eye Hospital, Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
14
|
Nassrallah WB, Li HR, Irani L, Wijesinghe P, Hogg PW, Hui L, Oh J, Mackenzie IR, Hirsch-Reinshagen V, Hsiung GYR, Pham W, Lee S, Matsubara JA. 3-Dimensional morphological characterization of neuroretinal microglia in Alzheimer's disease via machine learning. Acta Neuropathol Commun 2024; 12:202. [PMID: 39719599 DOI: 10.1186/s40478-024-01898-6] [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: 08/18/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024] Open
Abstract
Alzheimer's Disease (AD) is a debilitating neurodegenerative disease that affects 47.5 million people worldwide. AD is characterised by the formation of plaques containing extracellular amyloid-β (Aβ) and neurofibrillary tangles composed of hyper-phosphorylated tau proteins (pTau). Aβ gradually accumulates in the brain up to 20 years before the clinical onset of dementia, making it a compelling candidate for early detection of AD. It has been shown that there is increased deposition of Aβs in AD patients' retinas. However, little is known about microglia's ability to function and clear Aβ within the retina of AD and control eyes. We labelled microglia with ionised calcium-binding adaptor molecule 1 (IBA-1) in AD and age-matched control donor retinas. We then used interactive machine learning to segment individual microglia in 3D. In the temporal mid-peripheral region, we found that the number of microglia was significantly lower in AD retinas compared to controls. Unexpectedly, the size of the microglia was significantly larger in the AD retinas compared to controls. We also labelled retinal microglia for Cluster of Differentiation 68 (CD68), a transmembrane glycoprotein expressed by cells in the monocyte lineage and a marker of phagocytic activity and activated microglia. The size of CD68 + cells was statistically different between AD and control microglial, with CD68 + cells being larger in AD. In contrast, there was no difference in either size or shape for CD68- microglia between the two groups, suggesting an important difference in the active states of CD68 + microglia in AD retina. There was also significantly increased CD68 immunoreactivity in individual microglia within the AD group. Overall, this study reveals unique differences in the size and activity of the retinal microglia, which may relate to their potential chronic activation due to increased levels of Aβs in the AD retina.
Collapse
Affiliation(s)
- Wissam B Nassrallah
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Hao Ran Li
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Lyden Irani
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Printha Wijesinghe
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Peter William Hogg
- Department of Cellular and Physiological Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Lucy Hui
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Jean Oh
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada
| | - Ian R Mackenzie
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Veronica Hirsch-Reinshagen
- Department of Pathology and Laboratory Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Ging-Yuek Robin Hsiung
- Division of Neurology, Department of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Wellington Pham
- Vanderbilt University School of Medicine, Vanderbilt University Institute of Imaging Science, Nashville, TN, USA
| | - Sieun Lee
- Simon Fraser University School of Engineering Science, Burnaby, BC, Canada
- Mental Health and Clinical Neurosciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Joanne A Matsubara
- Department of Ophthalmology and Visual Sciences, The University of British Columbia, 2550 Willow St. Room 375, Vancouver, BC, V5Z 3N9, Canada.
| |
Collapse
|
15
|
Van Gaever F, Mingneau F, Vanherle S, Driege Y, Haegman M, Van Wonterghem E, Xie J, Vandenbroucke RE, Hendriks JJA, Beyaert R, Staal J. The phytohormone abscisic acid enhances remyelination in mouse models of multiple sclerosis. Front Immunol 2024; 15:1500697. [PMID: 39742273 PMCID: PMC11685095 DOI: 10.3389/fimmu.2024.1500697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/27/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Over the past few decades, there has been a sudden rise in the incidence of Multiple Sclerosis (MS) in Western countries. However, current treatments often show limited efficacy in certain patients and are associated with adverse effects, which highlights the need for safer and more effective therapeutic approaches. Environmental factors, particularly dietary habits, have been observed to play a substantial role in the development of MS. In this study, we are the first to investigate the potential protective effect of the phytohormone abscisic acid (ABA) in MS. ABA, which is abundant in fruits such as figs, apricots and bilberries, is known to cross the blood-brain barrier and has demonstrated neuroprotective effects in conditions like depression and Alzheimer's disease. Methods In this study, we investigated whether ABA supplementation enhances remyelination in both ex vivo and in vivo mouse models. Results Our results indicated that ABA enhanced remyelination and that this enhanced remyelination is associated with increased lipid droplet load, reduced levels of degraded myelin, and a higher abundance of F4/80+ cells in the demyelinated brain of mice treated with ABA. In in vitro models, we further demonstrated that ABA treatment elevates lipid droplet formation by enhancing the phagocytic capacity of macrophages. Additionally, in a mouse model of microglial activation, we showed that ABA-treated mice maintain a less inflammatory microglial phenotype. Conclusion Our findings highlight a crucial role for macrophages and microglia in enabling ABA to enhance the remyelination process. Furthermore, ABA's ability to improve remyelination together with its ability to reduce microglial activation, make ABA a promising candidate for modulating macrophage phenotype and reducing neuroinflammation in MS.
Collapse
Affiliation(s)
- Femke Van Gaever
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Fleur Mingneau
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Sam Vanherle
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Yasmine Driege
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Mira Haegman
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Elien Van Wonterghem
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Junhua Xie
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Roosmarijn E. Vandenbroucke
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jerome J. A. Hendriks
- Department of Immunology and Infection, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- University MS Center Hasselt, Pelt, Belgium
| | - Rudi Beyaert
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Jens Staal
- VIB-UGent Center for Inflammation Research, VIB, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
- Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| |
Collapse
|
16
|
Chu CT, Uruno A, Katsuoka F, Yamamoto M. Role of NRF2 in Pathogenesis of Alzheimer's Disease. Antioxidants (Basel) 2024; 13:1529. [PMID: 39765857 PMCID: PMC11727090 DOI: 10.3390/antiox13121529] [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: 10/18/2024] [Revised: 12/09/2024] [Accepted: 12/10/2024] [Indexed: 01/15/2025] Open
Abstract
Alzheimer's disease (AD) is a polygenic, multifactorial neurodegenerative disorder and remains the most prevalent form of dementia, globally. Despite decades of research efforts, there is still no effective cure for this debilitating condition. AD research has increasingly focused on transcription factor NRF2 (nuclear factor erythroid 2-related factor 2) as a potential therapeutic target. NRF2 plays a crucial role in protecting cells and tissues from environmental stressors, such as electrophiles and reactive oxygen species. Recently, an increasing number of studies have demonstrated that NRF2 is a key regulator in AD pathology. NRF2 is highly expressed in microglia, resident macrophages in the central nervous system, and contributes to neuroinflammation, phagocytosis and neurodegeneration in AD. NRF2 has been reported to modulate microglia-induced inflammation and facilitate the transition from homeostatic microglia to a disease-associated microglia subset. Genetic and pharmacological activation of NRF2 has been demonstrated to improve cognitive function. Here, we review the current understanding of the involvement of NRF2 in AD and the critical role that NRF2 plays in microglia in the context of AD. Our aim is to highlight the potential of targeting NRF2 in the microglia as a promising therapeutic strategy for mitigating the progression of AD.
Collapse
Affiliation(s)
- Ching-Tung Chu
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| | - Akira Uruno
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| | - Fumiki Katsuoka
- Department of Integrative Genomics, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan;
- Advanced Research Center for Innovations in Next-Generation Medicine, Tohoku University, Sendai 980-8573, Japan
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai 980-8573, Japan; (C.-T.C.); (A.U.)
| |
Collapse
|
17
|
Maya-Arteaga JP, Martínez-Orozco H, Diaz-Cintra S. MorphoGlia, an interactive method to identify and map microglia morphologies, demonstrates differences in hippocampal subregions of an Alzheimer's disease mouse model. Front Cell Neurosci 2024; 18:1505048. [PMID: 39698052 PMCID: PMC11653188 DOI: 10.3389/fncel.2024.1505048] [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: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/20/2024] Open
Abstract
Microglia are dynamic central nervous system cells crucial for maintaining homeostasis and responding to neuroinflammation, as evidenced by their varied morphologies. Existing morphology analysis often fails to detect subtle variations within the full spectrum of microglial morphologies due to their reliance on predefined categories. Here, we present MorphoGlia, an interactive, user-friendly pipeline that objectively characterizes microglial morphologies. MorphoGlia employs a machine learning ensemble to select relevant morphological features of microglia cells, perform dimensionality reduction, cluster these features, and subsequently map the clustered cells back onto the tissue, providing a spatial context for the identified microglial morphologies. We applied this pipeline to compare the responses between saline solution (SS) and scopolamine (SCOP) groups in a SCOP-induced mouse model of Alzheimer's disease, with a specific focus on the hippocampal subregions CA1 and Hilus. Next, we assessed microglial morphologies across four groups: SS-CA1, SCOP-CA1, SS-Hilus, and SCOP-Hilus. The results demonstrated that MorphoGlia effectively differentiated between SS and SCOP-treated groups, identifying distinct clusters of microglial morphologies commonly associated with pro-inflammatory states in the SCOP groups. Additionally, MorphoGlia enabled spatial mapping of these clusters, identifying the most affected hippocampal layers. This study highlights MorphoGlia's capability to provide unbiased analysis and clustering of microglial morphological states, making it a valuable tool for exploring microglial heterogeneity and its implications for central nervous system pathologies.
Collapse
Affiliation(s)
| | | | - Sofía Diaz-Cintra
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Santiago de Querétaro, Mexico
| |
Collapse
|
18
|
Steffens S, Mäkinen H, Stenberg T, Wigren HK. Microglial morphology aligns with vigilance stage-specific neuronal oscillations in a brain region-dependent manner. Glia 2024; 72:2344-2356. [PMID: 39301843 DOI: 10.1002/glia.24617] [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: 05/30/2024] [Revised: 08/21/2024] [Accepted: 08/26/2024] [Indexed: 09/22/2024]
Abstract
Microglia, the resident immune cells in the brain, dynamically adapt their morphology based on their functional state. This study explored the relationship between microglial morphology and sleep-wake cycles in mice. Using Iba1 immunostaining to identify microglia, we quantified morphological changes in microglia at different timepoints in multiple brain regions (cortex, hippocampus, basal forebrain, hindbrain, and cerebellum) in B6 male mice using semi-automated 3D structural analysis. Simultaneously, in a separate group, we monitored wake and sleep stage-specific brain activity using EEG/EMG recordings. During natural sleep-wake cycles, we observed increased microglial complexity (enlarged volume, territorial coverage, and ramification) during wakefulness, characterized by high-frequency theta (8-12 Hz) and gamma activity (30-80 Hz). Conversely, during NREM sleep, which is dominated by delta activity (0.5-4 Hz), microglia displayed reduced complexity. Notably, this pattern was absent in brain regions lacking direct functional connections to areas generating vigilance stage-dependent thalamocortical oscillations. We then extended wakefulness to decouple circadian influence from sleep-wake-specific neuronal activity. This procedure attenuated the decrease in microglial complexity observed during natural sleep, suggesting a crucial role for neuronal activity. Subsequent recovery sleep restored microglial features, independent of the time of day (zeitgeber time). These findings reveal a dynamic interplay between vigilance stage-specific thalamocortical activity and microglial morphology across various brain regions. This suggests a potential role for microglia in sleep regulation and warrants further investigation to understand the underlying mechanisms.
Collapse
Affiliation(s)
- Sarah Steffens
- SLEEPWELL Research Program I Faculty of Medicine, University of Helsinki, Finland
| | - Hilla Mäkinen
- SLEEPWELL Research Program I Faculty of Medicine, University of Helsinki, Finland
| | - Tarja Stenberg
- SLEEPWELL Research Program I Faculty of Medicine, University of Helsinki, Finland
| | - Henna-Kaisa Wigren
- SLEEPWELL Research Program I Faculty of Medicine, University of Helsinki, Finland
- Molecular and Integrative Biosciences Research Programme I Faculty of Biological and Environmental Sciences, University of Helsinki, Finland
| |
Collapse
|
19
|
Latini L, De Araujo DSM, Amato R, Canovai A, Buccarello L, De Logu F, Novelli E, Vlasiuk A, Malerba F, Arisi I, Florio R, Asari H, Capsoni S, Strettoi E, Villetti G, Imbimbo BP, Dal Monte M, Nassini R, Geppetti P, Marinelli S, Cattaneo A. A p75 neurotrophin receptor-sparing nerve growth factor protects retinal ganglion cells from neurodegeneration by targeting microglia. Br J Pharmacol 2024; 181:4890-4919. [PMID: 39252503 DOI: 10.1111/bph.17316] [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: 01/16/2024] [Revised: 05/22/2024] [Accepted: 06/10/2024] [Indexed: 09/11/2024] Open
Abstract
BACKGROUND AND PURPOSE Retinal ganglion cells (RGCs) are the output stage of retinal information processing, via their axons forming the optic nerve (ON). ON damage leads to axonal degeneration and death of RGCs, and results in vision impairment. Nerve growth factor (NGF) signalling is crucial for RGC operations and visual functions. Here, we investigate a new neuroprotective mechanism of a novel therapeutic candidate, a p75-less, TrkA-biased NGF agonist (hNGFp) in rat RGC degeneration, in comparison with wild type human NGF (hNGFwt). EXPERIMENTAL APPROACH Both neonate and adult rats, whether subjected or not to ON lesion, were treated with intravitreal injections or eye drops containing either hNGFp or hNGFwt. Different doses of the drugs were administered at days 1, 4 or 7 after injury for a maximum of 10 days, when immunofluorescence, electrophysiology, cellular morphology, cytokine array and behaviour studies were carried out. Pharmacokinetic evaluation was performed on rabbits treated with hNGFp ocular drops. RESULTS hNGFp exerted a potent RGC neuroprotection by acting on microglia cells, and outperformed hNGFwt in rescuing RGC degeneration and reducing inflammatory molecules. Delayed use of hNGFp after ON lesion resulted in better outcomes compared with treatment with hNGFwt. Moreover, hNGFp-based ocular drops were less algogenic than hNGFwt. Pharmacokinetic measurements revealed that biologically relevant quantities of hNGFp were found in the rabbit retina. CONCLUSIONS AND IMPLICATIONS Our data point to microglia as a new cell target through which NGF-induced TrkA signalling exerts neuroprotection of the RGC, emphasizing hNGFp as a powerful treatment to tackle retinal degeneration.
Collapse
Affiliation(s)
- Laura Latini
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | | | - Rosario Amato
- Department of Biology, University of Pisa, Pisa, Italy
| | | | - Lucia Buccarello
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Francesco De Logu
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Elena Novelli
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Anastasiia Vlasiuk
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
- Epigenetics and Neurobiology Unit, EMBL Rome, European Molecular Biology Laboratory, Rome, Italy
| | - Francesca Malerba
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Ivan Arisi
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Rita Florio
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Hiroki Asari
- Faculty of Biosciences, Collaboration for Joint PhD Degree Between EMBL and Heidelberg University, Heidelberg, Germany
| | - Simona Capsoni
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
- Section of Human Physiology, Department of Neuroscience and Rehabilitation, University of Ferrara, Ferrara, Italy
| | - Enrica Strettoi
- Institute of Neuroscience, Italian National Research Council-CNR, Pisa, Italy
| | - Gino Villetti
- Department of Research & Development, Chiesi Farmaceutici, Parma, Italy
| | | | | | - Romina Nassini
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Pierangelo Geppetti
- Department of Health Sciences, Clinical Pharmacology and Oncology Section, University of Florence, Florence, Italy
| | - Silvia Marinelli
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
| | - Antonino Cattaneo
- European Brain Research Institute-Fondazione Rita Levi-Montalcini, Rome, Italy
- BIO@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy
| |
Collapse
|
20
|
Butler B, Renney M, Bennett K, Charpentier G, Nance E. A rotenone organotypic whole hemisphere slice model of mitochondrial abnormalities in the neonatal brain. J Biol Eng 2024; 18:67. [PMID: 39543609 PMCID: PMC11566268 DOI: 10.1186/s13036-024-00465-w] [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: 07/12/2024] [Accepted: 10/30/2024] [Indexed: 11/17/2024] Open
Abstract
Mitochondrial abnormalities underscore a variety of neurologic injuries and diseases and are well-studied in adult populations. Clinical studies identify critical roles of mitochondria in a wide range of developmental brain injuries, but models that capture mitochondrial abnormalities in systems representative of the neonatal brain environment are lacking. Here, we develop an organotypic whole-hemisphere (OWH) brain slice model of mitochondrial dysfunction in the neonatal brain. We extended the utility of complex I inhibitor rotenone (ROT), canonically used in models of adult neurodegenerative diseases, to inflict mitochondrial damage in OWH slices from term-equivalent rats. We quantified whole-slice health over 6 days of exposure for a range of doses represented in ROT literature. We identified 50 nM ROT as a suitable exposure level for OWH slices to inflict injury without compromising viability. At the selected exposure level, we confirmed exposure- and time-dependent mitochondrial responses showing differences in mitochondrial fluorescence and nuclear localization using MitoTracker imaging in live OWH slices and dysregulated mitochondrial markers via RT-qPCR screening. We leveraged the regional structures present in OWH slices to quantify cell density and cell death in the cortex and the midbrain regions, observing higher susceptibilities to damage in the midbrain as a function of exposure and culture time. We supplemented these findings with analysis of microglia and mature neurons showing time-, region-, and exposure-dependent differences in microglial responses. We demonstrated changes in tissue microstructure as a function of region, culture time, and exposure level using live-video epifluorescence microscopy of extracellularly diffusing nanoparticle probes in live OWH slices. Our results highlight severity-, time-, and region-dependent responses and establish a complimentary model system of mitochondrial abnormalities for high-throughput or live-tissue experimental needs.
Collapse
Affiliation(s)
- Brendan Butler
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Malcolm Renney
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Kristin Bennett
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Gisele Charpentier
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA
| | - Elizabeth Nance
- Department of Chemical Engineering, University of Washington, Seattle, WA, 98195, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, 98195, USA.
- Molecular Engineering & Sciences Institute, University of Washington, Seattle, WA, 98195, USA.
| |
Collapse
|
21
|
Li Q, Xie Y, Lin J, Li M, Gu Z, Xin T, Zhang Y, Lu Q, Guo Y, Xing Y, Wang W. Microglia Sing the Prelude of Neuroinflammation-Associated Depression. Mol Neurobiol 2024:10.1007/s12035-024-04575-w. [PMID: 39535682 DOI: 10.1007/s12035-024-04575-w] [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: 06/14/2023] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Major depressive disorder (MDD) is a psychiatric condition characterized by sadness and anhedonia and is closely linked to chronic low-grade neuroinflammation, which is primarily induced by microglia. Nonetheless, the mechanisms by which microglia elicit depressive symptoms remain uncertain. This review focuses on the mechanism linking microglia and depression encompassing the breakdown of the blood-brain barrier, the hypothalamic-pituitary-adrenal axis, the gut-brain axis, the vagus and sympathetic nervous systems, and the susceptibility influenced by epigenetic modifications on microglia. These pathways may lead to the alterations of microglia in cytokine levels, as well as increased oxidative stress. Simultaneously, many antidepressant treatments can alter the immune phenotype of microglia, while anti-inflammatory treatments can also have antidepressant effects. This framework linking microglia, neuroinflammation, and depression could serve as a reference for targeting microglia to treat depression.
Collapse
Affiliation(s)
- Qingqing Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Ying Xie
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Jinyi Lin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Miaomiao Li
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Ziyan Gu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Tianli Xin
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yang Zhang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Qixia Lu
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yihui Guo
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China
| | - Yanhong Xing
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| | - Wuyang Wang
- Jiangsu Province Key Laboratory of Anesthesiology, Jiangsu Province Key Laboratory of Anesthesia and Analgesia Application Technology, NMPA Key Laboratory for Research and Evaluation of Narcotic and Psychotropic Drugs, Xuzhou Medical University, 209 Tongshan Rd, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
22
|
Lyubomudrov M, Babkina A, Tsokolaeva Z, Yadgarov M, Shigeev S, Sundukov D, Golubev A. Morphology of Cortical Microglia in the Hyperacute Phase of Subarachnoid Hemorrhage. BIOLOGY 2024; 13:917. [PMID: 39596872 PMCID: PMC11591589 DOI: 10.3390/biology13110917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024]
Abstract
Hemorrhagic stroke is the deadliest type of stroke. Cellular and molecular biomarkers are important for understanding the pathophysiology of stroke. Microglia are among the most promising biological markers. However, the morphological and physiological characteristics of microglia, as well as the structural and functional aspects of their interactions with neurons and other cells, are largely unknown. Due to the large number of different morphological phenotypes and very limited information on microglial changes in subarachnoid hemorrhage (SAH), we performed this study aimed at identifying the features of the distribution of various microglial phenotypes in the layers of the cerebral cortex in the hyperacute phase of non-traumatic SAH. We studied the distribution of various microglial phenotypes in the layers of the cerebral cortex of SAH non-survivors with a control group (coronary heart disease and sudden cardiac death were the underlying causes of death). An immunohistochemical study using antibodies to iba-1 (a marker of microglia) revealed changes in the morphological phenotypes of microglia in the cerebral cortex after subarachnoid hemorrhage. Significant differences between the groups indicate a rapid microglial response to injury. The findings indicate that there are quantitative and phenotypic changes in microglia in the cerebral cortex during early SAH in the human cortex.
Collapse
Affiliation(s)
- Maksim Lyubomudrov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Anastasiya Babkina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Zoya Tsokolaeva
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Mikhail Yadgarov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Sergey Shigeev
- Bureau of Forensic Medical Examination of the Department of Healthcare of the City of Moscow, Moscow 115516, Russia
| | - Dmitriy Sundukov
- Institute of Medicine, Peoples’ Friendship University of Russia Named after Patrice Lumumba, Moscow 117198, Russia
| | - Arkady Golubev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
- Institute of Medicine, Peoples’ Friendship University of Russia Named after Patrice Lumumba, Moscow 117198, Russia
| |
Collapse
|
23
|
Liu Y, Cai X, Shi B, Mo Y, Zhang J, Luo W, Yu B, Li X. Mechanisms and Therapeutic Prospects of Microglia-Astrocyte Interactions in Neuropathic Pain Following Spinal Cord Injury. Mol Neurobiol 2024:10.1007/s12035-024-04562-1. [PMID: 39470872 DOI: 10.1007/s12035-024-04562-1] [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: 05/24/2024] [Accepted: 10/16/2024] [Indexed: 11/01/2024]
Abstract
Neuropathic pain is a prevalent and debilitating condition experienced by the majority of individuals with spinal cord injury (SCI). The complex pathophysiology of neuropathic pain, involving continuous activation of microglia and astrocytes, reactive gliosis, and altered neuronal plasticity, poses significant challenges for effective treatment. This review focuses on the pivotal roles of microglia and astrocytes, the two major glial cell types in the central nervous system, in the development and maintenance of neuropathic pain after SCI. We highlight the extensive bidirectional interactions between these cells, mediated by the release of inflammatory mediators, neurotransmitters, and neurotrophic factors, which contribute to the amplification of pain signaling. Understanding the microglia-astrocyte crosstalk and its impact on neuronal function is crucial for developing novel therapeutic strategies targeting neuropathic pain. In addition, this review discusses the fundamental biology, post-injury pain roles, and therapeutic prospects of microglia and astrocytes in neuropathic pain after SCI and elucidates the specific signaling pathways involved. We also speculated that the extracellular matrix (ECM) can affect the glial cells as well. Furthermore, we also mentioned potential targeted therapies, challenges, and progress in clinical trials, as well as new biomarkers and therapeutic targets. Finally, other relevant cell interactions in neuropathic pain and the role of glial cells in other neuropathic pain conditions have been discussed. This review serves as a comprehensive resource for further investigations into the microglia-astrocyte interaction and the detailed mechanisms of neuropathic pain after SCI, with the aim of improving therapeutic efficacy.
Collapse
Affiliation(s)
- Yinuo Liu
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xintong Cai
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bowen Shi
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Yajie Mo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Jianmin Zhang
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Wenting Luo
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Bodong Yu
- The Clinical Medical College, Nanchang University, Nanchang, 330006, Jiangxi, China
| | - Xi Li
- Department of Anesthesiology, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
24
|
Lin D, Kaye S, Chen M, Lyanna A, Ye L, Hammond LA, Gao J. Transcriptome and proteome profiling reveals TREM2-dependent and -independent glial response and metabolic perturbation in an Alzheimer's mouse model. J Biol Chem 2024; 300:107874. [PMID: 39395805 PMCID: PMC11570940 DOI: 10.1016/j.jbc.2024.107874] [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: 03/15/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/14/2024] Open
Abstract
Elucidating the intricate molecular mechanisms of Alzheimer's disease (AD) requires a multidimensional analysis incorporating various omics data. In this study, we employed transcriptome and proteome profiling of AppNL-G-F, a human APP knock-in model of amyloidosis, at the early and mid-stages of amyloid-beta (Aβ) pathology to delineate the impacts of Aβ deposition on brain cells. By contrasting AppNL-G-F mice with TREM2 (Triggering receptor expressed on myeloid cells 2) knockout models, our study further investigates the role of TREM2, a well-known AD risk gene, in influencing microglial responses to Aβ pathology. Our results highlight altered microglial states as a central feature of Aβ pathology, characterized by the significant upregulation of microglia-specific genes related to immune responses such as complement system and antigen presentation, and catabolic pathways such as phagosome formation and lysosome biogenesis. The absence of TREM2 markedly diminishes the induction of these genes, impairs Aβ clearance, and exacerbates dystrophic neurite formation. Importantly, TREM2 is required for the microglial engagement with Aβ plaques and the formation of compact Aβ plaque cores. Furthermore, this study reveals substantial disruptions in energy metabolism and protein synthesis, signaling a shift from anabolism to catabolism in response to Aβ deposition. This metabolic alteration, coupled with a decrease in synaptic protein abundance, occurs independently of TREM2, suggesting the direct effects of Aβ deposition on synaptic integrity and plasticity. In summary, our findings demonstrate altered microglial states and metabolic disruption following Aβ deposition, offering mechanistic insights into Aβ pathology and highlighting the potential of targeting these pathways in AD therapy.
Collapse
Affiliation(s)
- Da Lin
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Sarah Kaye
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Min Chen
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Amogh Lyanna
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Lihua Ye
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Luke A Hammond
- Department of Neurology, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA
| | - Jie Gao
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, Ohio, USA.
| |
Collapse
|
25
|
Wellford SA, Chen CW, Vukovic M, Batich KA, Lin E, Shalek AK, Ordovas-Montanes J, Moseman AP, Moseman EA. Distinct olfactory mucosal macrophage populations mediate neuronal maintenance and pathogen defense. Mucosal Immunol 2024; 17:1102-1113. [PMID: 39074615 PMCID: PMC11483463 DOI: 10.1016/j.mucimm.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/27/2024] [Accepted: 07/26/2024] [Indexed: 07/31/2024]
Abstract
The olfactory mucosa is important for both the sense of smell and as a mucosal immune barrier to the upper airway and brain. However, little is known about how the immune system mediates the conflicting goals of neuronal maintenance and inflammation in this tissue. A number of immune cell populations reside within the olfactory mucosa and yet we have little understanding of how these resident olfactory immune cells functionally interact with the chemosensory environment. Identifying these interactions will allow therapeutic manipulations that treat disorders such as post-viral olfactory dysfunction. Macrophages are the most prevalent immune cell type in the uninflamed olfactory mucosa and here, we identify two distinct tissue macrophage populations in murine olfactory mucosa. P2ry12hi macrophages are transcriptionally specialized for neuron interactions, closely associated with olfactory neuron cell bodies, long-term tissue residents, and functionally specialized to phagocytose cells and debris, including olfactory neurons. Conversely, MHC Class IIhi macrophages are transcriptionally dedicated to cytokine production and antigen presentation, localized primarily within the olfactory lamina propria, more rapidly replaced by blood monocytes, and rapidly produce chemokines in response to viral infection. We further show that these macrophage signatures are present in human olfactory biopsies, and P2ry12-like olfactory macrophages are reduced in patients with long-term smell loss following COVID-19. Together, these data show that two olfactory macrophage populations regulate neurons and initiate the immune response, contributing to our understanding of both olfactory immunity and tissue-resident macrophage biology.
Collapse
Affiliation(s)
- Sebastian A Wellford
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States; Cell Signaling and Immunity Section, Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ching-Wen Chen
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
| | - Marko Vukovic
- Broad Institute of MIT and Harvard, Cambridge, MA, United States; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States; Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA, United States; Department of Chemistry, MIT, Cambridge, MA, USA
| | - Kristen A Batich
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States; Department of Neurosurgery, Duke University School of Medicine, Durham, NC, United States
| | - Elliot Lin
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
| | - Alex K Shalek
- Broad Institute of MIT and Harvard, Cambridge, MA, United States; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States; Institute for Medical Engineering and Science (IMES), MIT, Cambridge, MA, United States; Department of Chemistry, MIT, Cambridge, MA, USA; Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Jose Ordovas-Montanes
- Broad Institute of MIT and Harvard, Cambridge, MA, United States; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, United States; Division of Gastroenterology, Hepatology, and Nutrition, Boston Children's Hospital, Boston, MA, United States; Harvard Stem Cell Institute, Cambridge, MA, United States; Program in Immunology, Harvard Medical School, Boston, MA, United States
| | - Annie Park Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States
| | - E Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, United States.
| |
Collapse
|
26
|
Kim J, Sullivan O, Lee K, Jao J, Tamayo J, Madany AM, Wong B, Ashwood P, Ciernia AV. Repeated LPS induces training and tolerance of microglial responses across brain regions. J Neuroinflammation 2024; 21:233. [PMID: 39304952 PMCID: PMC11414187 DOI: 10.1186/s12974-024-03198-1] [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: 04/08/2024] [Accepted: 08/08/2024] [Indexed: 09/22/2024] Open
Abstract
BACKGROUND Neuroinflammation is involved in the pathogenesis of almost every central nervous system disorder. As the brain's innate immune cells, microglia fine tune their activity to a dynamic brain environment. Previous studies have shown that repeated bouts of peripheral inflammation can trigger long-term changes in microglial gene expression and function, a form of innate immune memory. METHODS AND RESULTS In this study, we used multiple low-dose lipopolysaccharide (LPS) injections in adult mice to study the acute cytokine, transcriptomic, and microglia morphological changes that contribute to the formation of immune memory in the frontal cortex, hippocampus, and striatum, as well as the long-term effects of these changes on behavior. Training and tolerance of gene expression was shared across regions, and we identified 3 unique clusters of DEGs (2xLPS-sensitive, 4xLPS-sensitive, LPS-decreased) enriched for different biological functions. 2xLPS-sensitive DEG promoters were enriched for binding sites for IRF and NFkB family transcription factors, two key regulators of innate immune memory. We quantified shifts in microglia morphological populations and found that while the proportion of ramified and rod-like microglia mostly remained consistent within brain regions and sexes with LPS treatment, there was a shift from ameboid towards hypertrophic morphological states across immune memory states and a dynamic emergence and resolution of events of microglia aligning end-to-end with repeated LPS. CONCLUSIONS Together, findings support the dynamic regulation of microglia during the formation of immune memories in the brain and support future work to exploit this model in brain disease contexts.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Justin Jao
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Juan Tamayo
- MIND Institute, University of California Davis, Davis, USA
| | | | - Brandon Wong
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada
| | - Paul Ashwood
- MIND Institute, University of California Davis, Davis, USA
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
- Djavad Mowafaghian Centre for Brain Health, Vancouver, Canada.
| |
Collapse
|
27
|
Theophanous S, Sargiannidou I, Kleopa KA. Glial Cells as Key Regulators in Neuroinflammatory Mechanisms Associated with Multiple Sclerosis. Int J Mol Sci 2024; 25:9588. [PMID: 39273535 PMCID: PMC11395575 DOI: 10.3390/ijms25179588] [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: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.
Collapse
Affiliation(s)
- Styliani Theophanous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
28
|
Fyke Z, Johansson R, Scott AI, Wiley D, Chelsky D, Zak JD, Al Nakouzi N, Koster KP, Yoshii A. Reduction of neuroinflammation and seizures in a mouse model of CLN1 batten disease using the small molecule enzyme mimetic, N-Tert-butyl hydroxylamine. Mol Genet Metab 2024; 143:108537. [PMID: 39033629 PMCID: PMC11473239 DOI: 10.1016/j.ymgme.2024.108537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/11/2024] [Accepted: 07/12/2024] [Indexed: 07/23/2024]
Abstract
Infantile neuronal ceroid lipofuscinosis (CLN1 Batten Disease) is a devastating pediatric lysosomal storage disease caused by pathogenic variants in the CLN1 gene, which encodes the depalmitoylation enzyme, palmitoyl-protein thioesterase 1 (PPT1). CLN1 patients present with visual deterioration, psychomotor dysfunction, and recurrent seizures until neurodegeneration results in death, typically before fifteen years of age. Histopathological features of CLN1 include aggregation of lysosomal autofluorescent storage material (AFSM), as well as profound gliosis. The current management of CLN1 is relegated to palliative care. Here, we examine the therapeutic potential of a small molecule PPT1 mimetic, N-tert-butyl hydroxylamine (NtBuHA), in a Cln1-/- mouse model. Treatment with NtBuHA reduced AFSM accumulation both in vitro and in vivo. Importantly, NtBuHA treatment in Cln1-/- mice reduced neuroinflammation, mitigated epileptic episodes, and normalized motor function. Live cell imaging of Cln1-/- primary cortical neurons treated with NtBuHA partially rescued aberrant synaptic calcium dynamics, suggesting a potential mechanism contributing to the therapeutic effects of NtBuHA in vivo. Taken together, our findings provide supporting evidence for NtBuHA as a potential treatment for CLN1 Batten Disease.
Collapse
Affiliation(s)
- Zach Fyke
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States of America
| | - Rachel Johansson
- School of Medicine, University of California Davis, Sacramento, CA, United States of America; Circumvent Pharmaceuticals, Portland, OR, United States of America
| | - Anna I Scott
- Circumvent Pharmaceuticals, Portland, OR, United States of America; Department of Laboratories, Seattle Children's Hospital, Seattle, WA, United States of America
| | - Devin Wiley
- Circumvent Pharmaceuticals, Portland, OR, United States of America
| | - Daniel Chelsky
- Circumvent Pharmaceuticals, Portland, OR, United States of America
| | - Joseph D Zak
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, United States of America; Department of Psychology University of Illinois at Chicago, Chicago, IL, United States of America
| | - Nader Al Nakouzi
- Circumvent Pharmaceuticals, Portland, OR, United States of America.
| | - Kevin P Koster
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States of America; Department of Neurobiology, University of Chicago, Chicago, IL, United States of America.
| | - Akira Yoshii
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, IL, United States of America; Department of Pediatrics, University of Illinois at Chicago, Chicago, IL, United States of America; Department of Neurology, University of Illinois at Chicago, Chicago, IL, United States of America
| |
Collapse
|
29
|
King DP, Abdalaziz M, Majewska AK, Cameron JL, Fudge JL. Microglia morphology in the developing primate amygdala and effects of early life stress. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.608133. [PMID: 39211183 PMCID: PMC11360906 DOI: 10.1101/2024.08.15.608133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
A unique pool of immature glutamatergic neurons in the primate amygdala, known as the paralaminar nucleus (PL), are maturing between infancy and adolescence. The PL is a potential substrate for the steep growth curve of amygdala volume during this developmental period. A microglial component is also embedded among the PL neurons, and likely supports local neuronal maturation and emerging synaptogenesis. Microglia may alter neuronal growth following environmental perturbations such as stress. Using multiple measures, we first found that microglia in the infant primate PL had relatively large somas, and a small arbor size. In contrast, microglia in the adolescent PL had a smaller soma, and a larger dendritic arbor. We then examined microglial morphology in the PL after a novel maternal separation protocol, to examine the effects of early life stress. After maternal separation, the microglia had increased soma size, arbor size and complexity. Surprisingly, strong effects were seen not only in the infant PL, but also in the adolescent PL from subjects who had experienced the separation many years earlier. We conclude that under maternal-rearing conditions, PL microglia morphology tracks PL neuronal growth, progressing to a more 'mature' phenotype by adolescence. Maternal separation has long-lasting effects on microglia, altering their normal developmental trajectory, and resulting in a 'hyper-ramified' phenotype that persists for years. We speculate that these changes have consequences for neuronal development in young primates. Significance Statement The paralaminar (PL) nucleus of the amygdala is an important source of plasticity, due to its unique repository of immature glutamatergic neurons. PL immature neurons mature between birth and adolescence. This process is likely supported by synaptogenesis, which requires microglia. Between infancy and adolescence in macaques, PL microglia became more dense, and shifted to a 'ramified' phenotype, consistent with increased synaptic pruning functions. Early life stress in the form of maternal separation, however, blunted this normal trajectory, leading to persistent 'parainflammatory' microglial morphologies. We speculate that early life stress may alter PL neuronal maturation and synapse formation through microglia.
Collapse
|
30
|
Gan YL, Lin WJ, Fang YC, Tang CY, Lee YH, Jeng CJ. FKBP51 is involved in LPS-induced microglial activation via NF-κB signaling to mediate neuroinflammation. Life Sci 2024; 351:122867. [PMID: 38914303 DOI: 10.1016/j.lfs.2024.122867] [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: 04/27/2024] [Revised: 06/03/2024] [Accepted: 06/20/2024] [Indexed: 06/26/2024]
Abstract
AIMS FKBP5 encodes FKBP51, which has been implicated in stress-related psychiatric disorders, and its expression is often increased under chronic stress, contributing to mental dysfunctions. However, the precise role of FKBP51 in brain inflammation remains unclear. This study aimed to investigate the role of FKBP51 in microglia-mediated inflammatory responses in the central nervous system. MAIN METHODS We employed a peripheral lipopolysaccharide (LPS) administration model to compare microglial activation and cytokine gene expression between Fkbp5 knockout (Fkbp5-KO) and wild-type (WT) male mice. Additionally, we used both BV2 and primary microglia in vitro to examine how Fkbp5 deletion influenced inflammation-related pathways and microglial functions. KEY FINDINGS This study revealed that systemic LPS-induced microglial activation was significantly attenuated in Fkbp5-KO mice compared with WT mice. In Fkbp5-KO mice following the LPS challenge, there was a notable decrease in the expression of pro-inflammatory genes, coupled with an increase in the anti-inflammatory gene Arg1. Furthermore, Fkbp5 knockdown in BV2 microglial cells led to reduced expression of LPS-induced inflammatory markers, and targeted inhibition of NF-κB activation, while Akt signaling remained unaffected. Similar results were observed in Fkbp5-KO primary microglia, which exhibited not only decreased microglial activation but also a significant reduction in phagocytic activity in response to LPS stimulation. SIGNIFICANCE This study highlights the critical role of FKBP51 in LPS-induced microglial activation and neuroinflammation. It shows that reducing FKBP51 levels attenuates inflammation through NF-κB signaling in microglia. This suggests that FKBP51 is a potential target for alleviating neuroinflammation-induced stress responses.
Collapse
Affiliation(s)
- Yu-Ling Gan
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wan-Jung Lin
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Department of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Ya-Ching Fang
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Department of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Chih-Yung Tang
- Department of Physiology, College of Medicine, National Taiwan University, Taipei 100, Taiwan
| | - Yi-Hsuan Lee
- Department and Institute of Physiology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| | - Chung-Jiuan Jeng
- Institute of Anatomy and Cell Biology, College of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei 112, Taiwan.
| |
Collapse
|
31
|
Vilca SJ, Margetts AV, Höglund L, Fleites I, Bystrom LL, Pollock TA, Bourgain-Guglielmetti F, Wahlestedt C, Tuesta LM. Microglia contribute to methamphetamine reinforcement and reflect persistent transcriptional and morphological adaptations to the drug. Brain Behav Immun 2024; 120:339-351. [PMID: 38838836 PMCID: PMC11269013 DOI: 10.1016/j.bbi.2024.05.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 06/07/2024] Open
Abstract
Methamphetamine use disorder (MUD) is a chronic, relapsing disease that is characterized by repeated drug use despite negative consequences and for which there are currently no FDA-approved cessation therapeutics. Repeated methamphetamine (METH) use induces long-term gene expression changes in brain regions associated with reward processing and drug-seeking behavior, and recent evidence suggests that methamphetamine-induced neuroinflammation may also shape behavioral and molecular responses to the drug. Microglia, the resident immune cells in the brain, are principal drivers of neuroinflammatory responses and contribute to the pathophysiology of substance use disorders. Here, we investigated transcriptional and morphological changes in dorsal striatal microglia in response to methamphetamine-taking and during methamphetamine abstinence, as well as their functional contribution to drug-taking behavior. We show that methamphetamine self-administration induces transcriptional changes associated with protein folding, mRNA processing, immune signaling, and neurotransmission in dorsal striatal microglia. Importantly, many of these transcriptional changes persist through abstinence, a finding supported by morphological analyses. Functionally, we report that microglial ablation increases methamphetamine-taking, possibly involving neuroimmune and neurotransmitter regulation. In contrast, microglial depletion during abstinence does not alter methamphetamine-seeking. Taken together, these results suggest that methamphetamine induces both short and long-term changes in dorsal striatal microglia that contribute to altered drug-taking behavior and may provide valuable insights into the pathophysiology of MUD.
Collapse
Affiliation(s)
- Samara J Vilca
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Alexander V Margetts
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Leon Höglund
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Isabella Fleites
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Lauren L Bystrom
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Tate A Pollock
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Florence Bourgain-Guglielmetti
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States
| | - Luis M Tuesta
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136, United States; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, United States.
| |
Collapse
|
32
|
Yang D, Sun Y, Lin D, Li S, Zhang Y, Wu A, Wei C. Interleukin-33 ameliorates perioperative neurocognitive disorders by modulating microglial state. Neuropharmacology 2024; 253:109982. [PMID: 38701943 DOI: 10.1016/j.neuropharm.2024.109982] [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: 12/14/2023] [Revised: 04/16/2024] [Accepted: 04/30/2024] [Indexed: 05/06/2024]
Abstract
Perioperative neurocognitive disorders (PND) are cognitive dysfunctions that usually occur in elderly patients after anesthesia and surgery. Microglial overactivation is a key underlying mechanism. Interleukin-33 (IL-33) is a member of the IL-1 family that orchestrates microglial function. In the present study, we explored how IL-33, which regulates microglia, contributes to cognitive improvement in a male mouse model of PND. An exploratory laparotomy was performed to establish a PND model. The expression levels of IL-33 and its receptor ST2 were evaluated using Western blot. IL-33/ST2 secretion, microglial density, morphology, phagocytosis of synapse, and proliferation, and dystrophic microglia were assessed using immunofluorescence. Synaptic plasticity was measured using Golgi staining and long-term potentiation. The Morris water maze and open field test were used to evaluate cognitive function and anxiety. Hippocampal expression of IL-33 and ST2 were elevated on postoperative day 3. We confirmed that IL-33 was secreted by astrocytes and neurons, whereas ST2 mainly colocalized with microglia. IL-33 treatment induced microgliosis after anesthesia and surgery. These microglia had larger soma sizes and shorter and fragmented branches. Compared to the Surgery group, IL-33 treatment reduced the synaptic phagocytosis of microglia and increased microglial proliferation and dystrophic microglia. IL-33 treatment also reversed the impaired synaptic plasticity and cognitive function caused by anesthesia and surgery. In conclusion, these results indicate that IL-33 plays a key role in regulating microglial state and synaptic phagocytosis in a PND mouse model. IL-33 treatment has a therapeutic potential for improving cognitive dysfunction in PND.
Collapse
Affiliation(s)
- Di Yang
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yi Sun
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Dandan Lin
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Yan Zhang
- State Key Laboratory of Membrane Biology, College of Life Sciences, Peking University, Beijing, China.
| | - Anshi Wu
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| | - Changwei Wei
- Department of Anesthesiology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
33
|
Frechou MA, Martin SS, McDermott KD, Huaman EA, Gökhan Ş, Tomé WA, Coen-Cagli R, Gonçalves JT. Adult neurogenesis improves spatial information encoding in the mouse hippocampus. Nat Commun 2024; 15:6410. [PMID: 39080283 PMCID: PMC11289285 DOI: 10.1038/s41467-024-50699-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: 12/15/2022] [Accepted: 06/24/2024] [Indexed: 08/02/2024] Open
Abstract
Adult neurogenesis is a unique form of neuronal plasticity in which newly generated neurons are integrated into the adult dentate gyrus in a process that is modulated by environmental stimuli. Adult-born neurons can contribute to spatial memory, but it is unknown whether they alter neural representations of space in the hippocampus. Using in vivo two-photon calcium imaging, we find that male and female mice previously housed in an enriched environment, which triggers an increase in neurogenesis, have increased spatial information encoding in the dentate gyrus. Ablating adult neurogenesis blocks the effect of enrichment and lowers spatial information, as does the chemogenetic silencing of adult-born neurons. Both ablating neurogenesis and silencing adult-born neurons decreases the calcium activity of dentate gyrus neurons, resulting in a decreased amplitude of place-specific responses. These findings are in contrast with previous studies that suggested a predominantly inhibitory action for adult-born neurons. We propose that adult neurogenesis improves representations of space by increasing the gain of dentate gyrus neurons and thereby improving their ability to tune to spatial features. This mechanism may mediate the beneficial effects of environmental enrichment on spatial learning and memory.
Collapse
Affiliation(s)
- M Agustina Frechou
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Laboratory of Neurotechnology and Biophysics, The Rockefeller University, New York, NY, USA
| | - Sunaina S Martin
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Psychology, University of California San Diego, La Jolla, CA, USA
| | - Kelsey D McDermott
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Evan A Huaman
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Şölen Gökhan
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Wolfgang A Tomé
- Saul R. Korey Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Radiation Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ruben Coen-Cagli
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Systems and Computational Biology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, NY, USA
| | - J Tiago Gonçalves
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA.
- Gottesmann Institute for Stem Cell Biology and Regenerative Medicine, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
34
|
Darlot F, Villard P, Salam LA, Rousseau L, Piret G. Glial scarring around intra-cortical MEA implants with flexible and free microwires inserted using biodegradable PLGA needles. Front Bioeng Biotechnol 2024; 12:1408088. [PMID: 39104630 PMCID: PMC11298340 DOI: 10.3389/fbioe.2024.1408088] [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: 03/27/2024] [Accepted: 05/29/2024] [Indexed: 08/07/2024] Open
Abstract
Introduction: Many invasive and noninvasive neurotechnologies are being developed to help treat neurological pathologies and disorders. Making a brain implant safe, stable, and efficient in the long run is one of the requirements to conform with neuroethics and overcome limitations for numerous promising neural treatments. A main limitation is low biocompatibility, characterized by the damage implants create in brain tissue and their low adhesion to it. This damage is partly linked to friction over time due to the mechanical mismatch between the soft brain tissue and the more rigid wires. Methods: Here, we performed a short biocompatibility assessment of bio-inspired intra-cortical implants named "Neurosnooper" made of a microelectrode array consisting of a thin, flexible polymer-metal-polymer stack with microwires that mimic axons. Implants were assembled into poly-lactic-glycolic acid (PLGA) biodegradable needles for their intra-cortical implantation. Results and Discussion: The study of glial scars around implants, at 7 days and 2 months post-implantation, revealed a good adhesion between the brain tissue and implant wires and a low glial scar thickness. The lowest corresponds to electrode wires with a section size of 8 μm × 10 μm, compared to implants with the 8 μm × 50 μm electrode wire section size, and a straight shape appears to be better than a zigzag. Therefore, in addition to flexibility, size and shape parameters are important when designing electrode wires for the next generation of clinical intra-cortical implants.
Collapse
Affiliation(s)
- Fannie Darlot
- Braintech Laboratory, Institut National de la Santé et de la Recherche Médicale U1205, Université Grenoble Alpes, Grenoble, France
| | - Paul Villard
- Braintech Laboratory, Institut National de la Santé et de la Recherche Médicale U1205, Université Grenoble Alpes, Grenoble, France
| | - Lara Abdel Salam
- Braintech Laboratory, Institut National de la Santé et de la Recherche Médicale U1205, Université Grenoble Alpes, Grenoble, France
| | | | - Gaëlle Piret
- Braintech Laboratory, Institut National de la Santé et de la Recherche Médicale U1205, Université Grenoble Alpes, Grenoble, France
| |
Collapse
|
35
|
Carrier M, Robert MÈ, St-Pierre MK, Ibáñez FG, Gonçalves de Andrade E, Laroche A, Picard K, Vecchiarelli HA, Savage JC, Boilard É, Desjardins M, Tremblay MÈ. Bone marrow-derived myeloid cells transiently colonize the brain during postnatal development and interact with glutamatergic synapses. iScience 2024; 27:110037. [PMID: 39021809 PMCID: PMC11253522 DOI: 10.1016/j.isci.2024.110037] [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: 10/09/2023] [Revised: 03/01/2024] [Accepted: 05/16/2024] [Indexed: 07/20/2024] Open
Abstract
Although the roles of embryonic yolk sac-derived, resident microglia in neurodevelopment were extensively studied, the possible involvement of bone marrow-derived cells remains elusive. In this work, we used a fate-mapping strategy to selectively label bone marrow-derived cells and their progeny in the brain (FLT3+IBA1+). FLT3+IBA1+ cells were confirmed to be transiently present in the healthy brain during early postnatal development. FLT3+IBA1+ cells have a distinct morphology index at postnatal day(P)0, P7, and P14 compared with neighboring microglia. FLT3+IBA1+ cells also express the microglial markers P2RY12 and TMEM119 and interact with VGLUT1 synapses at P14. Scanning electron microscopy indeed showed that FLT3+ cells contact and engulf pre-synaptic elements. Our findings suggest FLT3+IBA1+ cells might assist microglia in their physiological functions in the developing brain including synaptic pruning which is performed using their purinergic sensors. Our findings stimulate further investigation on the involvement of peripheral macrophages during homeostatic and pathological development.
Collapse
Affiliation(s)
- Micaël Carrier
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Département de psychiatrie et de neurosciences, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
| | - Marie-Ève Robert
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Marie-Kim St-Pierre
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Fernando González Ibáñez
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | | | - Audrée Laroche
- Département de microbiologie et immunologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Katherine Picard
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
| | | | - Julie C. Savage
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
| | - Éric Boilard
- Département de microbiologie et immunologie, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
| | - Michèle Desjardins
- Department of Physics, Physical Engineering and Optics, Université Laval, Québec City, QC G1V 0A6, Canada
- Oncology Division, Centre de recherche du CHU de Québec, Université Laval, Québec City, QC G1V 4G2, Canada
| | - Marie-Ève Tremblay
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC G1V 4G2, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 3E6, Canada
- Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec, QC G1V 0A6, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC V6T 1Z4 Canada
- Department of Neurology and Neurosurgery, McGill University, Montréal, QC H3A 0G4 Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC V8W 2Y2, Canada
| |
Collapse
|
36
|
Kim J, Pavlidis P, Ciernia AV. Development of a High-Throughput Pipeline to Characterize Microglia Morphological States at a Single-Cell Resolution. eNeuro 2024; 11:ENEURO.0014-24.2024. [PMID: 39029952 PMCID: PMC11289588 DOI: 10.1523/eneuro.0014-24.2024] [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: 01/10/2024] [Revised: 04/29/2024] [Accepted: 05/07/2024] [Indexed: 07/21/2024] Open
Abstract
As rapid responders to their environments, microglia engage in functions that are mirrored by their cellular morphology. Microglia are classically thought to exhibit a ramified morphology under homeostatic conditions which switches to an ameboid form during inflammatory conditions. However, microglia display a wide spectrum of morphologies outside of this dichotomy, including rod-like, ramified, ameboid, and hypertrophic states, which have been observed across brain regions, neurodevelopmental timepoints, and various pathological contexts. We applied dimensionality reduction and clustering to consider contributions of multiple morphology measures together to define a spectrum of microglial morphological states in a mouse dataset that we used to demonstrate the utility of our toolset. Using ImageJ, we first developed a semiautomated approach to characterize 27 morphology features from hundreds to thousands of individual microglial cells in a brain region-specific manner. Within this pool of features, we defined distinct sets of highly correlated features that describe different aspects of morphology, including branch length, branching complexity, territory span, and circularity. When considered together, these sets of features drove different morphological clusters. Our tools captured morphological states similarly and robustly when applied to independent datasets and using different immunofluorescent markers for microglia. We have compiled our morphology analysis pipeline into an accessible, easy-to-use, and fully open-source ImageJ macro and R package that the neuroscience community can expand upon and directly apply to their own analyses. Outcomes from this work will supply the field with new tools to systematically evaluate the heterogeneity of microglia morphological states across various experimental models and research questions.
Collapse
Affiliation(s)
- Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, British Columbia V6T 1Z3, Canada
| | - Paul Pavlidis
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, British Columbia V6T 1Z3, Canada
- Department of Psychiatry, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Michael Smith Laboratories, Vancouver, British Columbia V6T 1Z4, Canada
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, British Columbia V6T 1Z4, Canada
- Djavad Mowafaghian Centre for Brain Health, Vancouver, British Columbia V6T 1Z3, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia V6T 1Z3, Canada
| |
Collapse
|
37
|
Boucher ML, Conley G, Morriss NJ, Ospina-Mora S, Qiu J, Mannix R, Meehan WP. Time-Dependent Long-Term Effect of Memantine following Repetitive Mild Traumatic Brain Injury. J Neurotrauma 2024; 41:e1736-e1758. [PMID: 38666723 DOI: 10.1089/neu.2023.0423] [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: 05/16/2024] Open
Abstract
Repetitive mild traumatic brain injury (rmTBI, e.g., sports concussions) may be associated with both acute and chronic symptoms and neurological changes. Despite the common occurrence of these injuries, therapeutic strategies are limited. One potentially promising approach is N-methyl-D-aspartate receptor (NMDAR) blockade to alleviate the effects of post-injury glutamatergic excitotoxicity. Initial pre-clinical work using the NMDAR antagonist, memantine, suggests that immediate treatment following rmTBI improves a variety of acute outcomes. It remains unclear (1) whether acute memantine treatment has long-term benefits and (2) whether delayed treatment following rmTBI is beneficial, which are both clinically relevant concerns. To test this, animals were subjected to rmTBI via a weight drop model with rotational acceleration (five hits in 5 days) and randomized to memantine treatment immediately, 3 months, or 6 months post-injury, with a treatment duration of one month. Behavioral outcomes were assessed at 1, 4, and 7 months post-injury. Neuropathological outcomes were characterized at 7 months post-injury. We observed chronic changes in behavior (anxiety-like behavior, motor coordination, spatial learning, and memory), as well as neuroinflammation (microglia, astrocytes) and tau phosphorylation (T231). Memantine treatment, either immediately or 6 months post-injury, appears to confer greater rescue of neuroinflammatory changes (microglia) than vehicle or treatment at the 3-month time point. Although memantine is already being prescribed chronically to address persistent symptoms associated with rmTBI, this study represents the first evidence of which we are aware to suggest a small but durable effect of memantine treatment in mild, concussive injuries. This effect suggests that memantine, although potentially beneficial, is insufficient to treat all aspects of rmTBI alone and should be combined with other therapeutic agents in a multi-therapy approach, with attention given to the timing of treatment.
Collapse
Affiliation(s)
- Masen L Boucher
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
| | | | - Nicholas J Morriss
- University of Rochester School of Medicine and Dentistry, University of Rochester Medical Center, Rochester, New York, USA
| | | | - Jianhua Qiu
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Rebekah Mannix
- Division of Emergency Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - William P Meehan
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
- Division of Sports Medicine, Boston Children's Hospital, Boston, Massachusetts, USA
- The Micheli Center for Sports Injury Prevention, Waltham, Massachusetts, USA
| |
Collapse
|
38
|
Diniz DG, de Oliveira JHP, Guerreiro LCF, de Menezes GC, de Assis ACL, Duarte TQ, dos Santos IBD, Maciel FD, Soares GLDS, Araújo SC, Franco FTDC, do Carmo EL, Morais RDAB, de Lima CM, Brites D, Anthony DC, Diniz JAP, Diniz CWP. Contrasting Disease Progression, Microglia Reactivity, Tolerance, and Resistance to Toxoplasma gondii Infection in Two Mouse Strains. Biomedicines 2024; 12:1420. [PMID: 39061995 PMCID: PMC11274029 DOI: 10.3390/biomedicines12071420] [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: 04/18/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 07/28/2024] Open
Abstract
Our study investigated the innate immune response to Toxoplasma gondii infection by assessing microglial phenotypic changes and sickness behavior as inflammatory response markers post-ocular tachyzoite instillation. Disease progression in Swiss albino mice was compared with the previously documented outcomes in BALB/c mice using an identical ocular route and parasite burden (2 × 105 tachyzoites), with saline as the control. Contrary to expectations, the Swiss albino mice displayed rapid, lethal disease progression, marked by pronounced sickness behaviors and mortality within 11-12 days post-infection, while the survivors exhibited no apparent signs of infection. Comparative analysis revealed the T. gondii-infected BALB/c mice exhibited reduced avoidance of feline odors, while the infected Swiss albino mice showed enhanced avoidance responses. There was an important increase in microglial cells in the dentate gyrus molecular layer of the infected Swiss albino mice compared to the BALB/c mice and their respective controls. Hierarchical cluster and discriminant analyses identified three microglial morphological clusters, differentially affected by T. gondii infection across strains. The BALB/c mice exhibited increased microglial branching and complexity, while the Swiss albino mice showed reduced shrunken microglial arbors, diminishing their morphological complexity. These findings highlight strain-specific differences in disease progression and inflammatory regulation, indicating lineage-specific mechanisms in inflammatory responses, tolerance, and resistance. Understanding these elements is critical in devising control measures for toxoplasmosis.
Collapse
Affiliation(s)
- Daniel G. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
- Núcleo de Pesquisas em Oncologia, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66075-110, Pará, Brazil
| | - Jhonnathan H. P. de Oliveira
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Luma C. F. Guerreiro
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Biologia Molecular e Neuroecologia, Instituto Federal do Pará, Campus Bragança, Bragança 68600-000, Pará, Brazil
| | - Gabriel C. de Menezes
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Alexa C. L. de Assis
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Tainá Q. Duarte
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Izabelly B. D. dos Santos
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Flávia D. Maciel
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Gabrielly L. da S. Soares
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| | - Sanderson C. Araújo
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Felipe T. de C. Franco
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Ediclei L. do Carmo
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Rafaela dos A. B. Morais
- Seção de Parasitologia, Instituto Evandro Chagas, Belém 67030-000, Pará, Brazil; (E.L.d.C.); (R.d.A.B.M.)
| | - Camila M. de Lima
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Dora Brites
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal;
- Department of Pharmaceutical Sciences and Medicines, Faculty of Pharmacy, Universidade de Lisboa, 1649-003 Lisbon, Portugal
| | - Daniel C. Anthony
- Laboratory of Experimental Neuropathology, Department of Pharmacology, University of Oxford, Oxford OX1 2JD, UK;
| | - José A. P. Diniz
- Laboratório de Microscopia Eletrônica, Instituto Evandro Chagas, Belém 66077-830, Pará, Brazil; (S.C.A.); (F.T.d.C.F.); (J.A.P.D.)
| | - Cristovam W. P. Diniz
- Laboratório de Investigações em Neurodegeneração e Infecção, Instituto de Ciências Biológicas, Hospital Universitário João de Barros Barreto, Universidade Federal do Pará, Belém 66073-005, Pará, Brazil; (D.G.D.); (J.H.P.d.O.); (L.C.F.G.); (G.C.d.M.); (A.C.L.d.A.); (T.Q.D.); (I.B.D.d.S.); (F.D.M.); (G.L.d.S.S.); (C.M.d.L.)
| |
Collapse
|
39
|
Höfs L, Geißler-Lösch D, Wunderlich KM, Szegö EM, Van den Haute C, Baekelandt V, Hoyer W, Falkenburger BH. Evaluation of the Effect of β-Wrapin AS69 in a Mouse Model Based on Alpha-Synuclein Overexpression. Biomolecules 2024; 14:756. [PMID: 39062470 PMCID: PMC11274363 DOI: 10.3390/biom14070756] [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: 05/01/2024] [Revised: 06/03/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Aggregation of the protein α-Synuclein (αSyn) is a hallmark of Parkinson's disease (PD), dementia with Lewy bodies (DLB) and multiple systems atrophy, and alleviating the extent of αSyn pathology is an attractive strategy against neurodegeneration. The engineered binding protein β-wrapin AS69 binds monomeric αSyn. AS69 reduces primary and secondary nucleation as well as fibril elongation in vitro. It also mitigates aSyn pathology in a mouse model based on intrastriatal injection of aSyn pre-formed fibrils (PFFs). Since the PFF-based model does not represent all aspects of PD, we tested here whether AS69 can reduce neurodegeneration resulting from αSyn overexpression. Human A53T-αSyn was overexpressed in the mouse Substantia nigra (SN) by using recombinant adeno-associated viral vector (rAAV). AS69 was also expressed by rAAV transduction. Behavioral tests and immunofluorescence staining were used as outcomes. Transduction with rAAV-αSyn resulted in αSyn pathology as reported by phospho-αSyn staining and caused degeneration of dopaminergic neurons in the SN. The co-expression of rAAV-AS69 did not reduce αSyn pathology or the degeneration of dopaminergic neurons. We conclude that αSyn monomer binding by rAAV-AS69 was insufficient to protect from aSyn pathology resulting from αSyn overexpression.
Collapse
Affiliation(s)
- Lennart Höfs
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany (D.G.-L.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 01307 Dresden, Germany
| | - David Geißler-Lösch
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany (D.G.-L.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 01307 Dresden, Germany
| | - Kristof M. Wunderlich
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany (D.G.-L.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 01307 Dresden, Germany
| | - Eva M. Szegö
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany (D.G.-L.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 01307 Dresden, Germany
| | - Chris Van den Haute
- Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Veerle Baekelandt
- Leuven Viral Vector Core, KU Leuven, 3000 Leuven, Belgium
- Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven Brain Institute, KU Leuven, 3000 Leuven, Belgium
| | - Wolfgang Hoyer
- Institut für Physikalische Biologie, Heinrich-Heine-Universität Düsseldorf, 40225 Düsseldorf, Germany
- Institute of Biological Information Processing (IBI-7), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany
| | - Björn H. Falkenburger
- Department of Neurology, Technische Universität Dresden, 01307 Dresden, Germany (D.G.-L.)
- Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), 01307 Dresden, Germany
| |
Collapse
|
40
|
Song C, Wang Z, Cao J, Dong Y, Chen Y. Neurotoxic mechanisms of mycotoxins: Focus on aflatoxin B1 and T-2 toxin. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2024; 356:124359. [PMID: 38866317 DOI: 10.1016/j.envpol.2024.124359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 06/04/2024] [Accepted: 06/09/2024] [Indexed: 06/14/2024]
Abstract
Aflatoxin B1 (AFB1) and T-2 toxin are commonly found in animal feed and stored grain, posing a serious threat to human and animal health. Mycotoxins can penetrate brain tissue by compromising the blood-brain barrier, triggering oxidative stress and neuroinflammation, and leading to oxidative damage and apoptosis of brain cells. The potential neurotoxic mechanisms of AFB1 and T-2 toxin were discussed by summarizing the relevant research reports from the past ten years. AFB1 and T-2 toxin cause neuronal damage in the cerebral cortex and hippocampus, leading to synaptic transmission dysfunction, ultimately impairing the nervous system function of the body. The toxic mechanism is related to excessive reactive oxygen species (ROS), oxidative stress, mitochondrial dysfunction, apoptosis, autophagy, and an exaggerated inflammatory response. After passing through the blood-brain barrier, toxins can directly affect glial cells, alter the activation state of microglia and astrocytes, thereby promoting brain inflammation, disrupting the blood-brain barrier, and influencing the synaptic transmission process. We discussed the diverse effects of various concentrations of toxins and different modes of exposure on neurotoxicity. In addition, toxins can also cross the placental barrier, causing neurotoxic symptoms in offspring, as demonstrated in various species. Our goal is to uncover the underlying mechanisms of the neurotoxicity of AFB1 and T-2 toxin and to provide insights for future research, including investigating the impact of mycotoxins on interactions between microglia and astrocytes.
Collapse
Affiliation(s)
- Chao Song
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing, 100193, China
| | - Zixu Wang
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing, 100193, China
| | - Jing Cao
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing, 100193, China
| | - Yulan Dong
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing, 100193, China
| | - Yaoxing Chen
- College of Veterinary Medicine, China Agricultural University, Haidian, Beijing, 100193, China.
| |
Collapse
|
41
|
Wellford SA, Moseman EA. Olfactory immunology: the missing piece in airway and CNS defence. Nat Rev Immunol 2024; 24:381-398. [PMID: 38097777 PMCID: PMC11560121 DOI: 10.1038/s41577-023-00972-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2023] [Indexed: 12/23/2023]
Abstract
The olfactory mucosa is a component of the nasal airway that mediates the sense of smell. Recent studies point to an important role for the olfactory mucosa as a barrier to both respiratory pathogens and to neuroinvasive pathogens that hijack the olfactory nerve and invade the CNS. In particular, the COVID-19 pandemic has demonstrated that the olfactory mucosa is an integral part of a heterogeneous nasal mucosal barrier critical to upper airway immunity. However, our insufficient knowledge of olfactory mucosal immunity hinders attempts to protect this tissue from infection and other diseases. This Review summarizes the state of olfactory immunology by highlighting the unique immunologically relevant anatomy of the olfactory mucosa, describing what is known of olfactory immune cells, and considering the impact of common infectious diseases and inflammatory disorders at this site. We will offer our perspective on the future of the field and the many unresolved questions pertaining to olfactory immunity.
Collapse
Affiliation(s)
- Sebastian A Wellford
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
| | - E Ashley Moseman
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
42
|
Green TRF, Rowe RK. Quantifying microglial morphology: an insight into function. Clin Exp Immunol 2024; 216:221-229. [PMID: 38456795 PMCID: PMC11097915 DOI: 10.1093/cei/uxae023] [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: 08/09/2023] [Revised: 01/17/2024] [Accepted: 03/06/2024] [Indexed: 03/09/2024] Open
Abstract
Microglia are specialized immune cells unique to the central nervous system (CNS). Microglia have a highly plastic morphology that changes rapidly in response to injury or infection. Qualitative and quantitative measurements of ever-changing microglial morphology are considered a cornerstone of many microglia-centric research studies. The distinctive morphological variations seen in microglia are a useful marker of inflammation and severity of tissue damage. Although a wide array of damage-associated microglial morphologies has been documented, the exact functions of these distinct morphologies are not fully understood. In this review, we discuss how microglia morphology is not synonymous with microglia function, however, morphological outcomes can be used to make inferences about microglial function. For a comprehensive examination of the reactive status of a microglial cell, both histological and genetic approaches should be combined. However, the importance of quality immunohistochemistry-based analyses should not be overlooked as they can succinctly answer many research questions.
Collapse
Affiliation(s)
- Tabitha R F Green
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, USA
| | - Rachel K Rowe
- Department of Integrative Physiology, The University of Colorado Boulder, Boulder, CO, USA
| |
Collapse
|
43
|
Krejcová LV, Bento-Torres J, Diniz DG, Pereira A, Batista-de-Oliveira M, de Morais AACL, Mendes-da-Silva RF, Abadie-Guedes R, dos Santos ÂA, Lima DS, Guedes RCA, Picanço-Diniz CW. Unraveling the Influence of Litter Size, Maternal Care, Exercise, and Aging on Neurobehavioral Plasticity and Dentate Gyrus Microglia Dynamics in Male Rats. Brain Sci 2024; 14:497. [PMID: 38790475 PMCID: PMC11119659 DOI: 10.3390/brainsci14050497] [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: 04/11/2024] [Revised: 04/30/2024] [Accepted: 05/10/2024] [Indexed: 05/26/2024] Open
Abstract
This study explores the multifaceted influence of litter size, maternal care, exercise, and aging on rats' neurobehavioral plasticity and dentate gyrus microglia dynamics. Body weight evolution revealed a progressive increase until maturity, followed by a decline during aging, with larger litters exhibiting lower weights initially. Notably, exercised rats from smaller litters displayed higher body weights during the mature and aged stages. The dentate gyrus volumes showed no significant differences among groups, except for aged sedentary rats from smaller litters, which exhibited a reduction. Maternal care varied significantly based on litter size, with large litter dams showing lower frequencies of caregiving behaviors. Behavioral assays highlighted the detrimental impact of a sedentary lifestyle and reduced maternal care/large litters on spatial memory, mitigated by exercise in aged rats from smaller litters. The microglial dynamics in the layers of dentate gyrus revealed age-related changes modulated by litter size and exercise. Exercise interventions mitigated microgliosis associated with aging, particularly in aged rats. These findings underscore the complex interplay between early-life experiences, exercise, microglial dynamics, and neurobehavioral outcomes during aging.
Collapse
Affiliation(s)
- Lane Viana Krejcová
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - João Bento-Torres
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - Daniel Guerreiro Diniz
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
- Postgraduate Program in Oncology and Medical Sciences, João de Barros Barreto Universitary Hospital, Federal University of Pará, Belém 66075-110, Pará, Brazil
- Electron Microscopy Laboratory, Evandro Chagas Institute, Belém 66093-020, Pará, Brazil
| | - Antonio Pereira
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| | - Manuella Batista-de-Oliveira
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | | | | | - Ricardo Abadie-Guedes
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Ângela Amâncio dos Santos
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Denise Sandrelly Lima
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Rubem Carlos Araujo Guedes
- Naíde Teodósio Nutrition Physiology Laboratory, Department of Nutrition, Federal University of Pernambuco, Recife 50670-901, Pernambuco, Brazil
| | - Cristovam Wanderley Picanço-Diniz
- Neurodegeneration and Infection Research Laboratory, João de Barros Barreto Universitary Hospital, Institute of Biological Sciences, Federal University of Pará, Belém 66050-160, Pará, Brazil
| |
Collapse
|
44
|
Sheridan SD, Horng JE, Yeh H, McCrea L, Wang J, Fu T, Perlis RH. Loss of Function in the Neurodevelopmental Disease and Schizophrenia-Associated Gene CYFIP1 in Human Microglia-like Cells Supports a Functional Role in Synaptic Engulfment. Biol Psychiatry 2024; 95:676-686. [PMID: 37573007 PMCID: PMC10874584 DOI: 10.1016/j.biopsych.2023.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 07/18/2023] [Accepted: 07/23/2023] [Indexed: 08/14/2023]
Abstract
BACKGROUND The CYFIP1 gene, located in the neurodevelopmental risk locus 15q11.2, is highly expressed in microglia, but its role in human microglial function as it relates to neurodevelopment is not well understood. METHODS We generated multiple CRISPR (clustered regularly interspaced short palindromic repeat) knockouts of CYFIP1 in patient-derived models of microglia to characterize function and phenotype. Using microglia-like cells reprogrammed from peripheral blood mononuclear cells, we quantified phagocytosis of synaptosomes (isolated and purified synaptic vesicles) from human induced pluripotent stem cell (iPSC)-derived neuronal cultures as an in vitro model of synaptic pruning. We repeated these analyses in human iPSC-derived microglia-like cells derived from 3 isogenic wild-type/knockout line pairs derived from 2 donors and further characterized microglial development and function through morphology and motility. RESULTS CYFIP1 knockout using orthogonal CRISPR constructs in multiple patient-derived cell lines was associated with a statistically significant decrease in synaptic vesicle phagocytosis in microglia-like cell models derived from both peripheral blood mononuclear cells and iPSCs. Morphology was also shifted toward a more ramified profile, and motility was significantly reduced. However, iPSC-CYFIP1 knockout lines retained the ability to differentiate to functional microglia. CONCLUSIONS The changes in microglial phenotype and function due to the loss of function of CYFIP1 observed in this study implicate a potential impact on processes such as synaptic pruning that may contribute to CYFIP1-related neurodevelopmental disorders. Investigating risk genes in a range of central nervous system cell types, not solely neurons, may be required to fully understand the way in which common and rare variants intersect to yield neuropsychiatric disorders.
Collapse
Affiliation(s)
- Steven D Sheridan
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Joy E Horng
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Hana Yeh
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Liam McCrea
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Jennifer Wang
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Ting Fu
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts
| | - Roy H Perlis
- Center for Genomic Medicine and Department of Psychiatry, Massachusetts General Hospital, Boston, Massachusetts; Department of Psychiatry, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
45
|
Wei Z, Fang R, Wang Y, Dong J. Maternal exposure to di-(2-ethylhexyl) phthalate impaired the social interaction via activating microglia in male pups. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 272:116069. [PMID: 38340601 DOI: 10.1016/j.ecoenv.2024.116069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP), a common endocrine-disrupting chemical (EDC), is widely used in daily articles, early exposure to DEHP is associated with many behavioral changes in pups. This study aimed to investigate the effects and underlying mechanisms of maternal exposure to DEHP on the impaired social interaction in pups. Pregnant rats were administered 0, 30, 300, or 750 mg/kg/d DEHP daily by oral gavage. Highly aggressive proliferating immortalized (HAPI) cells were treated with mono-(2-ethylhexyl) phthalate (MEHP) and tyrosine phosphorylation inhibitor (AG490). Our results showed that DEHP exposure induced the activation of microglias (MGs) via activating the janus kinase 2 / signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway, and increased the level of pro-inflammatory factors, then impaired the social behavior in male pups, but not female pups. Moreover, MEHP exposure could also activate HAPI via activating this signaling pathway, and AG490 could inhibit the activation of this signaling pathway caused by MEHP. Therefore, we indicated that maternal exposure to DEHP could cause the gender-specific impaired social interaction in pups that might be related to the activation of MGs.
Collapse
Affiliation(s)
- Zhixia Wei
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, China
| | - Rui Fang
- Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, China
| | - Ying Wang
- Department of Obstetrics and Gynecology, Shengjing Hospital, China Medical University, 110004 Shenyang, China.
| | - Jing Dong
- Key Laboratory of Environmental Stress and Chronic Disease Control & Prevention (China Medical University), Ministry of Education, China Medical University, Shenyang, Liaoning 11012, China; Department of Occupational and Environmental Health, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang 110122, China.
| |
Collapse
|
46
|
Vilca SJ, Margetts AV, Fleites I, Wahlestedt C, Tuesta LM. Microglia contribute to methamphetamine reinforcement and reflect persistent transcriptional and morphological adaptations to the drug. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.19.563168. [PMID: 37961443 PMCID: PMC10634674 DOI: 10.1101/2023.10.19.563168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Methamphetamine use disorder (MUD) is a chronic, relapsing disease that is characterized by repeated drug use despite negative consequences and for which there are currently no FDA-approved cessation therapeutics. Repeated methamphetamine (METH) use induces long-term gene expression changes in brain regions associated with reward processing and drug-seeking behavior, and recent evidence suggests that methamphetamine-induced neuroinflammation may also shape behavioral and molecular responses to the drug. Microglia, the resident immune cells in the brain, are principal drivers of neuroinflammatory responses and contribute to the pathophysiology of substance use disorders. Here, we investigated transcriptional and morphological changes in dorsal striatal microglia in response to methamphetamine-taking and during methamphetamine abstinence, as well as their functional contribution to drug-taking behavior. We show that methamphetamine self-administration induces transcriptional changes associated with protein folding, mRNA processing, immune signaling, and neurotransmission in dorsal striatal microglia. Importantly, many of these transcriptional changes persist through abstinence, a finding supported by morphological analyses. Functionally, we report that microglial ablation increases methamphetamine-taking, possibly involving neuroimmune and neurotransmitter regulation, and that post-methamphetamine microglial repopulation attenuates drug-seeking following a 21-day period of abstinence. In contrast, microglial depletion during abstinence did not alter methamphetamine-seeking. Taken together, these results suggest that methamphetamine induces both short and long-term changes in dorsal striatal microglia that contribute to altered drug-taking behavior and may provide valuable insights into the pathophysiology of MUD.
Collapse
Affiliation(s)
- Samara J. Vilca
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Alexander V. Margetts
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Isabella Fleites
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Claes Wahlestedt
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| | - Luis M. Tuesta
- Department of Psychiatry & Behavioral Sciences, University of Miami Miller School of Medicine, Miami, FL 33136
- Center for Therapeutic Innovation, University of Miami Miller School of Medicine, Miami, FL 33136
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136
| |
Collapse
|
47
|
Carrier M, Hui CW, Watters V, Šimončičová E, Picard K, González Ibáñez F, Vernoux N, Droit A, Desjardins M, Tremblay MÈ. Behavioral as well as hippocampal transcriptomic and microglial responses differ across sexes in adult mouse offspring exposed to a dual genetic and environmental challenge. Brain Behav Immun 2024; 116:126-139. [PMID: 38016491 DOI: 10.1016/j.bbi.2023.11.025] [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: 02/27/2023] [Revised: 10/15/2023] [Accepted: 11/23/2023] [Indexed: 11/30/2023] Open
Abstract
INTRODUCTION A wide range of positive, negative, and cognitive symptoms compose the clinical presentation of schizophrenia. Schizophrenia is a multifactorial disorder in which genetic and environmental risk factors interact for a full emergence of the disorder. Infectious challenges during pregnancy are a well-known environmental risk factor for schizophrenia. Also, genetic variants affecting the function of fractalkine signaling between neurons and microglia were linked to schizophrenia. Translational animal models recapitulating these complex gene-environment associations have a great potential to untangle schizophrenia neurobiology and propose new therapeutic strategies. METHODS Given that genetic variants affecting the function of fractalkine signaling between neurons and microglia were linked to schizophrenia, we compared the outcomes of a well-characterized model of maternal immune activation induced using the viral mimetic polyinosinic:polycytidylic acid (Poly I:C) in wild-type versus fractalkine receptor knockout mice. Possible behavioral and immune alterations were assessed in male and female offspring during adulthood. Considering the role of the hippocampus in schizophrenia, microglial analyses and bulk RNA sequencing were performed within this region to assess the neuroimmune dynamics at play. Males and females were examined separately. RESULTS Offspring exposed to the dual challenge paradigm exhibited symptoms relevant to schizophrenia and unpredictably to mood disorders. Males displayed social and cognitive deficits related to schizophrenia, while females mainly presented anxiety-like behaviors related to mood disorders. Hippocampal microglia in females exposed to the dual challenge were hypertrophic, indicative of an increased surveillance, whereas those in males showed on the other end of the spectrum blunted morphologies with a reduced phagocytosis. Hippocampal bulk-RNA sequencing further revealed a downregulation in females of genes related to GABAergic transmission, which represents one of the main proposed causes of mood disorders. CONCLUSIONS Building on previous results, we identified in the current study distinctive behavioral phenotypes in female mice exposed to a dual genetic and environmental challenge, thus proposing a new model of neurodevelopmentally-associated mood and affective symptoms. This paves the way to future sex-specific investigations into the susceptibility to developmental challenges using animal models based on genetic and immune vulnerability as presented here.
Collapse
Affiliation(s)
- Micaël Carrier
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Psychiatry and Neuroscience, Faculty of Medicine, Université Laval, Québec City, QC, Canada; Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Chin W Hui
- Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Valérie Watters
- Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Eva Šimončičová
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Katherine Picard
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada; Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Fernando González Ibáñez
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada; Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Nathalie Vernoux
- Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
| | - Arnaud Droit
- Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada; Département de médecine moléculaire, Faculté de médecine, Université Laval, Québec City, QC, Canada
| | - Michèle Desjardins
- Department of Physics, Physical Engineering and Optics, Université Laval, Québec City, QC, Canada; Oncology Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
48
|
Traetta ME, Chaves Filho AM, Akinluyi ET, Tremblay MÈ. Neurodevelopmental and Neuropsychiatric Disorders. ADVANCES IN NEUROBIOLOGY 2024; 37:457-495. [PMID: 39207708 DOI: 10.1007/978-3-031-55529-9_26] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
This chapter will focus on microglial involvement in neurodevelopmental and neuropsychiatric disorders, particularly autism spectrum disorder (ASD), schizophrenia and major depressive disorder (MDD). We will describe the neuroimmune risk factors that contribute to the etiopathology of these disorders across the lifespan, including both in early life and adulthood. Microglia, being the resident immune cells of the central nervous system, could play a key role in triggering and determining the outcome of these disorders. This chapter will review preclinical and clinical findings where microglial morphology and function were examined in the contexts of ASD, schizophrenia and MDD. Clinical evidence points out to altered microglial morphology and reactivity, as well as increased expression of pro-inflammatory cytokines, supporting the idea that microglial abnormalities are involved in these disorders. Indeed, animal models for these disorders found altered microglial morphology and homeostatic functions which resulted in behaviours related to these disorders. Additionally, as microglia have emerged as promising therapeutic targets, we will also address in this chapter therapies involving microglial mechanisms for the treatment of neurodevelopmental and neuropsychiatric disorders.
Collapse
Affiliation(s)
| | | | - Elizabeth Toyin Akinluyi
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Pharmacology and Therapeutics, Afe Babalola University, Ado-Ekiti, Nigeria
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Département de Médecine Moléculaire, Université Laval, Quebec City, QC, Canada.
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Quebec City, QC, Canada.
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
- Institute on Aging and Lifelong Health (IALH), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
49
|
Visco DB, Manhães-de-Castro R, da Silva MM, Costa-de-Santana BJR, Pereira Dos Santos Junior J, Saavedra LM, de Lemos MDTB, Valdéz-Alarcón JJ, Lagranha CJ, Guzman-Quevedo O, Torner L, Toscano AE. Neonatal kaempferol exposure attenuates impact of cerebral palsy model on neuromotor development, cell proliferation, microglia activation, and antioxidant enzyme expression in the hippocampus of rats. Nutr Neurosci 2024; 27:20-41. [PMID: 36576161 DOI: 10.1080/1028415x.2022.2156034] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
OBJECTIVES This study aims to assess the effect of neonatal treatment with kaempferol on neuromotor development, proliferation of neural precursor cells, the microglia profile, and antioxidant enzyme gene expression in the hippocampus. METHODS A rat model of cerebral palsy was established using perinatal anoxia and sensorimotor restriction of hindlimbs during infancy. Kaempferol (1 mg/ kg) was intraperitoneally administered during the neonatal period. RESULTS Neonatal treatment with kaempferol reduces the impact of the cerebral palsy model on reflex ontogeny and on the maturation of physical features. Impairment of locomotor activity development and motor coordination was found to be attenuated by kaempferol treatment during the neonatal period in rats exposed to cerebral palsy. Neonatal treatment of kaempferol in cerebral palsy rats prevents a substantial reduction in the number of neural precursor cells in the dentate gyrus of the hippocampus, an activated microglia profile, and increased proliferation of microglia in the sub-granular zone and in the granular cell layer. Neonatal treatment with kaempferol increases gene expression of superoxide dismutase and catalase in the hippocampus of rats submitted to the cerebral palsy model. DISCUSSION Kaempferol attenuates the impact of cerebral palsy on neuromotor behavior development, preventing altered hippocampal microglia activation and mitigating impaired cell proliferation in a neurogenic niche in these rats. Neonatal treatment with kaempferol also increases antioxidant defense gene expression in the hippocampus of rats submitted to the cerebral palsy model.
Collapse
Affiliation(s)
- Diego Bulcão Visco
- Laboratory of Neurofunctional, Department of Biological Sciences and Health, Federal University of Amapá, Macapá, Brazil
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Raul Manhães-de-Castro
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Márcia Maria da Silva
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Bárbara J R Costa-de-Santana
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Joaci Pereira Dos Santos Junior
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
| | - Luís Miguel Saavedra
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | | | - Juan José Valdéz-Alarcón
- Centro Multidisciplinario de Estudios en Biotecnología - Facultad de Medicina Veterinaria y Zootecnia, Universidad Michoacana de San Nicolás de Hidalgo, Tarímbaro, Mexico
| | - Claudia Jacques Lagranha
- Graduate Program in Biochemistry and Physiology (PGBqF), Federal University of Pernambuco, Recife, Brazil
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Omar Guzman-Quevedo
- Instituto Tecnológico Superior de Tacámbaro, Tacámbaro, Mexico
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
| | - Luz Torner
- Centro de Investigación Biomédica de Michoacán, Instituto Mexicano del Seguro Social, Morelia, Mexico
| | - Ana Elisa Toscano
- Graduate Program in Nutrition (Posnutri), Health Sciences Center, Federal University of Pernambuco, Recife, Brazil
- Studies in Nutrition and Phenotypic Plasticity Unit, Department of Nutrition, Federal University of Pernambuco, Recife, Brazil
- Graduate Program in Neuropsychiatry and Behavioral Sciences (Posneuro), Federal University of Pernambuco, Recife, Brazil
- Nursing Unit, Vitória Academic Center, Federal University of Pernambuco, Vitória de Santo Antão, Brazil
| |
Collapse
|
50
|
Grewal S, Gonçalves de Andrade E, Kofoed RH, Matthews PM, Aubert I, Tremblay MÈ, Morse SV. Using focused ultrasound to modulate microglial structure and function. Front Cell Neurosci 2023; 17:1290628. [PMID: 38164436 PMCID: PMC10757935 DOI: 10.3389/fncel.2023.1290628] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 10/31/2023] [Indexed: 01/03/2024] Open
Abstract
Transcranial focused ultrasound (FUS) has the unique ability to target regions of the brain with high spatial precision, in a minimally invasive manner. Neuromodulation studies have shown that FUS can excite or inhibit neuronal activity, demonstrating its tremendous potential to improve the outcome of neurological diseases. Recent evidence has also shed light on the emerging promise that FUS has, with and without the use of intravenously injected microbubbles, in modulating the blood-brain barrier and the immune cells of the brain. As the resident immune cells of the central nervous system, microglia are at the forefront of the brain's maintenance and immune defense. Notably, microglia are highly dynamic and continuously survey the brain parenchyma by extending and retracting their processes. This surveillance activity aids microglia in performing key physiological functions required for brain activity and plasticity. In response to stressors, microglia rapidly alter their cellular and molecular profile to help facilitate a return to homeostasis. While the underlying mechanisms by which both FUS and FUS + microbubbles modify microglial structure and function remain largely unknown, several studies in adult mice have reported changes in the expression of the microglia/macrophage marker ionized calcium binding adaptor molecule 1, and in their phagocytosis, notably of protein aggregates, such as amyloid beta. In this review, we discuss the demonstrated and putative biological effects of FUS and FUS + microbubbles in modulating microglial activities, with an emphasis on the key cellular and molecular changes observed in vitro and in vivo across models of brain health and disease. Understanding how this innovative technology can modulate microglia paves the way for future therapeutic strategies aimed to promote beneficial physiological microglial roles, and prevent or treat maladaptive responses.
Collapse
Affiliation(s)
- Sarina Grewal
- Department of Bioengineering, Imperial College London, London, United Kingdom
- Department of Brain Sciences, Imperial College London, London, United Kingdom
| | - Elisa Gonçalves de Andrade
- Neuroscience Graduate Program, Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Rikke Hahn Kofoed
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Center for Experimental Neuroscience-CENSE, Department of Neurosurgery, Aarhus University Hospital, Aarhus, Denmark
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
| | - Paul M. Matthews
- Department of Brain Sciences, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| | - Isabelle Aubert
- Hurvitz Brain Sciences Research Program, Biological Sciences, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Sophie V. Morse
- Department of Bioengineering, Imperial College London, London, United Kingdom
- UK Dementia Research Institute, Imperial College London, London, United Kingdom
| |
Collapse
|