51
|
Xie M, Miller AS, Pallegar PN, Umpierre A, Liang Y, Wang N, Zhang S, Nagaraj NK, Fogarty ZC, Ghayal NB, Oskarsson B, Zhao S, Zheng J, Qi F, Nguyen A, Dickson DW, Wu LJ. Rod-shaped microglia interact with neuronal dendrites to regulate cortical excitability in TDP-43 related neurodegeneration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.30.601396. [PMID: 39005475 PMCID: PMC11244918 DOI: 10.1101/2024.06.30.601396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Motor cortical hyperexcitability is well-documented in the presymptomatic stage of amyotrophic lateral sclerosis (ALS). However, the mechanisms underlying this early dysregulation are not fully understood. Microglia, as the principal immune cells of the central nervous system, have emerged as important players in sensing and regulating neuronal activity. Here we investigated the role of microglia in the motor cortical circuits in a mouse model of TDP-43 neurodegeneration (rNLS8). Utilizing multichannel probe recording and longitudinal in vivo calcium imaging in awake mice, we observed neuronal hyperactivity at the initial stage of disease progression. Spatial and single-cell RNA sequencing revealed that microglia are the primary responders to motor cortical hyperactivity. We further identified a unique subpopulation of microglia, rod-shaped microglia, which are characterized by a distinct morphology and transcriptional profile. Notably, rod-shaped microglia predominantly interact with neuronal dendrites and excitatory synaptic inputs to attenuate motor cortical hyperactivity. The elimination of rod-shaped microglia through TREM2 deficiency increased neuronal hyperactivity, exacerbated motor deficits, and further decreased survival rates of rNLS8 mice. Together, our results suggest that rod-shaped microglia play a neuroprotective role by attenuating cortical hyperexcitability in the mouse model of TDP-43 related neurodegeneration.
Collapse
|
52
|
Pizzirusso G, Preka E, Goikolea J, Aguilar-Ruiz C, Rodriguez-Rodriguez P, Vazquez-Cabrera G, Laterza S, Latorre-Leal M, Eroli F, Blomgren K, Maioli S, Nilsson P, Fragkopoulou A, Fisahn A, Arroyo-García LE. Dynamic microglia alterations associate with hippocampal network impairments: A turning point in amyloid pathology progression. Brain Behav Immun 2024; 119:286-300. [PMID: 38608739 DOI: 10.1016/j.bbi.2024.04.009] [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: 11/21/2023] [Revised: 03/12/2024] [Accepted: 04/09/2024] [Indexed: 04/14/2024] Open
Abstract
Alzheimer's disease is a progressive neurological disorder causing memory loss and cognitive decline. The underlying causes of cognitive deterioration and neurodegeneration remain unclear, leading to a lack of effective strategies to prevent dementia. Recent evidence highlights the role of neuroinflammation, particularly involving microglia, in Alzheimer's disease onset and progression. Characterizing the initial phase of Alzheimer's disease can lead to the discovery of new biomarkers and therapeutic targets, facilitating timely interventions for effective treatments. We used the AppNL-G-F knock-in mouse model, which resembles the amyloid pathology and neuroinflammatory characteristics of Alzheimer's disease, to investigate the transition from a pre-plaque to an early plaque stage with a combined functional and molecular approach. Our experiments show a progressive decrease in the power of cognition-relevant hippocampal gamma oscillations during the early stage of amyloid pathology, together with a modification of fast-spiking interneuron intrinsic properties and postsynaptic input. Consistently, transcriptomic analyses revealed that these effects are accompanied by changes in synaptic function-associated pathways. Concurrently, homeostasis- and inflammatory-related microglia signature genes were downregulated. Moreover, we found a decrease in Iba1-positive microglia in the hippocampus that correlates with plaque aggregation and neuronal dysfunction. Collectively, these findings support the hypothesis that microglia play a protective role during the early stages of amyloid pathology by preventing plaque aggregation, supporting neuronal homeostasis, and overall preserving the oscillatory network's functionality. These results suggest that the early alteration of microglia dynamics could be a pivotal event in the progression of Alzheimer's disease, potentially triggering plaque deposition, impairment of fast-spiking interneurons, and the breakdown of the oscillatory circuitry in the hippocampus.
Collapse
Affiliation(s)
- Giusy Pizzirusso
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Sweden
| | - Efthalia Preka
- Department of Women's and Children's Health, Karolinska Institutet, Sweden
| | - Julen Goikolea
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Celia Aguilar-Ruiz
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Patricia Rodriguez-Rodriguez
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | | | - Simona Laterza
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Maria Latorre-Leal
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Francesca Eroli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Klas Blomgren
- Department of Women's and Children's Health, Karolinska Institutet, Sweden; Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Silvia Maioli
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden
| | | | - André Fisahn
- Department of Women's and Children's Health, Karolinska Institutet, Sweden.
| | - Luis Enrique Arroyo-García
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Karolinska Institutet, Sweden; Department of Women's and Children's Health, Karolinska Institutet, Sweden.
| |
Collapse
|
53
|
Zhang Y, Yan H, Wei Y, Wei X. Decoding mitochondria's role in immunity and cancer therapy. Biochim Biophys Acta Rev Cancer 2024; 1879:189107. [PMID: 38734035 DOI: 10.1016/j.bbcan.2024.189107] [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: 02/08/2024] [Revised: 04/22/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The functions of mitochondria, including energy production and biomolecule synthesis, have been known for a long time. Given the rising incidence of cancer, the role of mitochondria in cancer has become increasingly popular. Activated by components released by mitochondria, various pathways interact with each other to induce immune responses to protect organisms from attack. However, mitochondria play dual roles in the progression of cancer. Abnormalities in proteins, which are the elementary structures of mitochondria, are closely linked with oncogenesis. Both the aberrant accumulation of intermediates and mutations in enzymes result in the generation and progression of cancer. Therefore, targeting mitochondria to treat cancer may be a new strategy. Several drugs aimed at inhibiting mutated enzymes and accumulated intermediates have been tested clinically. Here, we discuss the current understanding of mitochondria in cancer and the interactions between mitochondrial functions, immune responses, and oncogenesis. Furthermore, we discuss mitochondria as hopeful targets for cancer therapy, providing insights into the progression of future therapeutic strategies.
Collapse
Affiliation(s)
- Yu Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Hong Yan
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 17, Block 3, Southern Renmin Road, 610041 Chengdu, Sichuan, PR China.
| |
Collapse
|
54
|
Chen R, Wang Z, Lin Q, Hou X, Jiang Y, Le Q, Liu X, Ma L, Wang F. Destabilization of fear memory by Rac1-driven engram-microglia communication in hippocampus. Brain Behav Immun 2024; 119:621-636. [PMID: 38670239 DOI: 10.1016/j.bbi.2024.04.024] [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: 09/21/2023] [Revised: 04/19/2024] [Accepted: 04/22/2024] [Indexed: 04/28/2024] Open
Abstract
Rac1 is a key regulator of the cytoskeleton and neuronal plasticity, and is known to play a critical role in psychological and cognitive brain disorders. To elucidate the engram specific Rac1 signaling in fear memory, a doxycycline (Dox)-dependent robust activity marking (RAM) system was used to label dorsal dentate gyrus (DG) engram cells in mice during contextual fear conditioning. Rac1 mRNA and protein levels in DG engram cells were peaked at 24 h (day 1) after fear conditioning and were more abundant in the fear engram cells than in the non-engram cells. Optogenetic activation of Rac1 in a temporal manner in DG engram cells before memory retrieval decreased the freezing level in the fear context. Optogenetic activation of Rac1 increased autophagy protein 7 (ATG7) expression in the DG engram cells and activated DG microglia. Microglia-specific transcriptomics and fluorescence in situ hybridization revealed that overexpression of ATG7 in the fear engram cells upregulated the mRNA of Toll-like receptor TLR2/4 in DG microglia. Knockdown of microglial TLR2/4 rescued fear memory destabilization induced by ATG7 overexpression or Rac1 activation in DG engram cells. These results indicate that Rac1-driven communications between engram cells and microglia contributes to contextual fear memory destabilization, and is mediated by ATG7 and TLR2/4, and suggest a novel mechanistic framework for the cytoskeletal regulator in fear memory interference.
Collapse
Affiliation(s)
- Ruyan Chen
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Zhilin Wang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qing Lin
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xutian Hou
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Yan Jiang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Qiumin Le
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Xing Liu
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Lan Ma
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China
| | - Feifei Wang
- School of Basic Medicine Sciences, State Key Laboratory of Medical Neurobiology, MOE Frontiers Center for Brain Science, Institutes of Brain Science, Pharmacology Research Center, Department of Neurology, Huashan Hospital, Fudan University, Shanghai 200032, China; Research Unit of Addition Memory, Chinese Academy of Medical Sciences (2021RU009), Shanghai 200032, China.
| |
Collapse
|
55
|
Berki P, Cserép C, Környei Z, Pósfai B, Szabadits E, Domonkos A, Kellermayer A, Nyerges M, Wei X, Mody I, Kunihiko A, Beck H, Kaikai H, Ya W, Lénárt N, Wu Z, Jing M, Li Y, Gulyás AI, Dénes Á. Microglia contribute to neuronal synchrony despite endogenous ATP-related phenotypic transformation in acute mouse brain slices. Nat Commun 2024; 15:5402. [PMID: 38926390 PMCID: PMC11208608 DOI: 10.1038/s41467-024-49773-1] [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: 12/30/2023] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Acute brain slices represent a workhorse model for studying the central nervous system (CNS) from nanoscale events to complex circuits. While slice preparation inherently involves tissue damage, it is unclear how microglia, the main immune cells and damage sensors of the CNS react to this injury and shape neuronal activity ex vivo. To this end, we investigated microglial phenotypes and contribution to network organization and functioning in acute brain slices. We reveal time-dependent microglial phenotype changes influenced by complex extracellular ATP dynamics through P2Y12R and CX3CR1 signalling, which is sustained for hours in ex vivo mouse brain slices. Downregulation of P2Y12R and changes of microglia-neuron interactions occur in line with alterations in the number of excitatory and inhibitory synapses over time. Importantly, functional microglia modulate synapse sprouting, while microglial dysfunction results in markedly impaired ripple activity both ex vivo and in vivo. Collectively, our data suggest that microglia are modulators of complex neuronal networks with important roles to maintain neuronal network integrity and activity. We suggest that slice preparation can be used to model time-dependent changes of microglia-neuron interactions to reveal how microglia shape neuronal circuits in physiological and pathological conditions.
Collapse
Affiliation(s)
- Péter Berki
- János Szentágothai Doctoral School of Neuroscience, Semmelweis University, Budapest, H-1083, Hungary
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Neuronal Network and Behaviour, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zsuzsanna Környei
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Eszter Szabadits
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Andor Domonkos
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
- Laboratory of Thalamus Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Anna Kellermayer
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Miklós Nyerges
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Xiaofei Wei
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Istvan Mody
- Department of Neurology, The David Geffen School of Medicine at UCLA, Los Angeles, CA, 90095, USA
| | - Araki Kunihiko
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - Heinz Beck
- Institute of Experimental Epileptology and Cognition Research, Medical University of Bonn, Bonn, 53127, Germany
- University Hospital Bonn, Bonn, Germany
| | - He Kaikai
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Wang Ya
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Zhaofa Wu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, 100101, Beijing, China
| | - Miao Jing
- Chinese Institute for Brain Research, 102206, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, New Cornerstone Science Laboratory, School of Life Sciences, Peking University, 100871, Beijing, China
| | - Attila I Gulyás
- Laboratory of Cerebral Cortex Research, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, HUN-REN Institute of Experimental Medicine, Budapest, H-1083, Hungary.
| |
Collapse
|
56
|
Campagno KE, Lu W, Sripinun P, Albalawi F, Cenaj A, Mitchell CH. Retinal microglial cells increase expression and release of IL-1β when exposed to ATP. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.25.600617. [PMID: 38979240 PMCID: PMC11230250 DOI: 10.1101/2024.06.25.600617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Cytokine IL-1β is an early component of inflammatory cascades, with both priming and activation steps required before IL-1β release. Here, the P2X7 receptor (P2X7R) for ATP was shown to both prime and release IL-1β from retinal microglial cells. Isolated retinal microglial cells increased expression of Il1b when stimulated with endogenous receptor agonist extracellular ATP; ATP also rapidly downregulated expression of microglial markers Tmem119 and Cd206. Changes to all three genes were reduced by specific P2X7R antagonist A839977, implicating the P2X7R. Microglial cells expressed the P2X7R on ramifications and responded to receptor agonist BzATP with robust and rapid rises in intracellular Ca 2+ . BzATP increased expression of IL-1β protein colocalizing with CX3CR1-GFP in retinal wholemounts consistent with microglial cells. ATP also triggered release of IL-1β from isolated retinal microglia into the bath; release was inhibited by A839977 and induced by BzATP, supporting a role for the P2X7R in release as well as priming. The IL-1β release triggered by ATP was substantially greater from microglial cells compared to astrocytes from the optic nerve head region. Il1b expression was increased by a transient rise in intraocular pressure and Il1b levels remained elevated 10 days after a single IOP elevation. In summary, this study suggests the P2X7 receptor can both prime IL-1β levels in microglial cells and trigger its release. The P2Y12R was previously identified as a chemoattractant for retinal microglia, suggesting the recruitment of the cells towards the source of released extracellular ATP could position microglia for P2X7R receptor, enabling both priming and release of IL-1β.
Collapse
|
57
|
Xi Y, Min Z, Liu M, Lin X, Yuan ZH. Role and recent progress of P2Y12 receptor in cancer development. Purinergic Signal 2024:10.1007/s11302-024-10027-w. [PMID: 38874752 DOI: 10.1007/s11302-024-10027-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: 03/05/2024] [Accepted: 06/03/2024] [Indexed: 06/15/2024] Open
Abstract
P2Y12 receptor (P2Y12R) is an adenosine-activated G protein-coupled receptor (GPCR) that plays a central role in platelet function, hemostasis, and thrombosis. P2Y12R activation can promote platelet aggregation and adhesion to cancer cells, promote tumor angiogenesis, and affect the tumor immune microenvironment (TIME) and tumor drug resistance, which is conducive to the progression of cancers. Meanwhile, P2Y12R inhibitors can inhibit this effect, suggesting that P2Y12R may be a potential therapeutic target for cancer. P2Y12R is involved in cancer development and metastasis, while P2Y12R inhibitors are effective in inhibiting cancer. However, a new study suggests that long-term use of P2Y12R inhibitors may increase the risk of cancer and the mechanism remains to be explored. In this paper, we reviewed the structural and functional characteristics of P2Y12R and its role in cancer. We explored the role of P2Y12R inhibitors in different tumors and the latest advances by summarizing the basic and clinical studies on the effects of P2Y12R inhibitors on tumors.
Collapse
Affiliation(s)
- Yanni Xi
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People's Republic of China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Zhenya Min
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Mianxue Liu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
- The Second Clinical Medical College, Nanchang University, Nanchang, 330006, Republic of China
| | - Xueqin Lin
- Department of Nursing, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, Republic of China
| | - Zhao-Hua Yuan
- Department of General Surgery, Jiujiang Hospital of Traditional Chinese Medicine, Jiujiang, Jiangxi, 332007, People's Republic of China.
| |
Collapse
|
58
|
Brennan FH, Swarts EA, Kigerl KA, Mifflin KA, Guan Z, Noble BT, Wang Y, Witcher KG, Godbout JP, Popovich PG. Microglia promote maladaptive plasticity in autonomic circuitry after spinal cord injury in mice. Sci Transl Med 2024; 16:eadi3259. [PMID: 38865485 DOI: 10.1126/scitranslmed.adi3259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 05/16/2024] [Indexed: 06/14/2024]
Abstract
Robust structural remodeling and synaptic plasticity occurs within spinal autonomic circuitry after severe high-level spinal cord injury (SCI). As a result, normally innocuous visceral or somatic stimuli elicit uncontrolled activation of spinal sympathetic reflexes that contribute to systemic disease and organ-specific pathology. How hyperexcitable sympathetic circuitry forms is unknown, but local cues from neighboring glia likely help mold these maladaptive neuronal networks. Here, we used a mouse model of SCI to show that microglia surrounded active glutamatergic interneurons and subsequently coordinated multi-segmental excitatory synaptogenesis and expansion of sympathetic networks that control immune, neuroendocrine, and cardiovascular functions. Depleting microglia during critical periods of circuit remodeling after SCI prevented maladaptive synaptic and structural plasticity in autonomic networks, decreased the frequency and severity of autonomic dysreflexia, and prevented SCI-induced immunosuppression. Forced turnover of microglia in microglia-depleted mice restored structural and functional indices of pathological dysautonomia, providing further evidence that microglia are key effectors of autonomic plasticity. Additional data show that microglia-dependent autonomic plasticity required expression of triggering receptor expressed on myeloid cells 2 (Trem2) and α2δ-1-dependent synaptogenesis. These data suggest that microglia are primary effectors of autonomic neuroplasticity and dysautonomia after SCI in mice. Manipulating microglia may be a strategy to limit autonomic complications after SCI or other forms of neurologic disease.
Collapse
Affiliation(s)
- Faith H Brennan
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Department of Biomedical and Molecular Sciences and Center for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Emily A Swarts
- Department of Biomedical and Molecular Sciences and Center for Neuroscience Studies, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Kristina A Kigerl
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Katherine A Mifflin
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Zhen Guan
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Benjamin T Noble
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Yan Wang
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Kristina G Witcher
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Jonathan P Godbout
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Phillip G Popovich
- Department of Neuroscience, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Belford Center for Spinal Cord Injury, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
- Institute for Behavioral Medicine Research, Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
59
|
Barclay KM, Abduljawad N, Cheng Z, Kim MW, Zhou L, Yang J, Rustenhoven J, Mazzitelli JA, Smyth LCD, Kapadia D, Brioschi S, Beatty W, Hou J, Saligrama N, Colonna M, Yu G, Kipnis J, Li Q. An inducible genetic tool to track and manipulate specific microglial states reveals their plasticity and roles in remyelination. Immunity 2024; 57:1394-1412.e8. [PMID: 38821054 PMCID: PMC11299637 DOI: 10.1016/j.immuni.2024.05.005] [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: 08/18/2023] [Revised: 02/14/2024] [Accepted: 05/07/2024] [Indexed: 06/02/2024]
Abstract
Recent single-cell RNA sequencing studies have revealed distinct microglial states in development and disease. These include proliferative-region-associated microglia (PAMs) in developing white matter and disease-associated microglia (DAMs) prevalent in various neurodegenerative conditions. PAMs and DAMs share a similar core gene signature. However, the extent of the dynamism and plasticity of these microglial states, as well as their functional significance, remains elusive, partly due to the lack of specific tools. Here, we generated an inducible Cre driver line, Clec7a-CreERT2, that targets PAMs and DAMs in the brain parenchyma. Utilizing this tool, we profiled labeled cells during development and in several disease models, uncovering convergence and context-dependent differences in PAM and DAM gene expression. Through long-term tracking, we demonstrated microglial state plasticity. Lastly, we specifically depleted DAMs in demyelination, revealing their roles in disease recovery. Together, we provide a versatile genetic tool to characterize microglial states in CNS development and disease.
Collapse
Affiliation(s)
- Kia M Barclay
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Nora Abduljawad
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Zuolin Cheng
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Min Woo Kim
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Immunology Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Lu Zhou
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jin Yang
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Justin Rustenhoven
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Jose A Mazzitelli
- Neuroscience Graduate Program, Washington University School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Medical Scientist Training Program, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Leon C D Smyth
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Dvita Kapadia
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Simone Brioschi
- Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - JinChao Hou
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Naresha Saligrama
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Department of Neurology, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA; Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine in St. Louis, St. Louis, MO 63112, USA
| | - Marco Colonna
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Guoqiang Yu
- Bradley Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Jonathan Kipnis
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA
| | - Qingyun Li
- Department of Neuroscience, Washington University in St. Louis School of Medicine, St. Louis, MO 63110, USA; Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, MO 63110, USA; Brain Immunology and Glia (BIG) Center, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine in St. Louis, School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
60
|
Huang X, Lee S, Chen K, Kawaguchi R, Wiskow O, Ghosh S, Frost D, Perrault L, Pandey R, Klim JR, Boivin B, Hermawan C, Livak KJ, Geschwind DH, Wainger BJ, Eggan KC, Bean BP, Woolf CJ. Downregulation of the silent potassium channel Kv8.1 increases motor neuron vulnerability in amyotrophic lateral sclerosis. Brain Commun 2024; 6:fcae202. [PMID: 38911266 PMCID: PMC11191651 DOI: 10.1093/braincomms/fcae202] [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: 08/08/2023] [Revised: 04/10/2024] [Accepted: 06/07/2024] [Indexed: 06/25/2024] Open
Abstract
While voltage-gated potassium channels have critical roles in controlling neuronal excitability, they also have non-ion-conducting functions. Kv8.1, encoded by the KCNV1 gene, is a 'silent' ion channel subunit whose biological role is complex since Kv8.1 subunits do not form functional homotetramers but assemble with Kv2 to modify its ion channel properties. We profiled changes in ion channel expression in amyotrophic lateral sclerosis patient-derived motor neurons carrying a superoxide dismutase 1(A4V) mutation to identify what drives their hyperexcitability. A major change identified was a substantial reduction of KCNV1/Kv8.1 expression, which was also observed in patient-derived neurons with C9orf72 expansion. We then studied the effect of reducing KCNV1/Kv8.1 expression in healthy motor neurons and found it did not change neuronal firing but increased vulnerability to cell death. A transcriptomic analysis revealed dysregulated metabolism and lipid/protein transport pathways in KCNV1/Kv8.1-deficient motor neurons. The increased neuronal vulnerability produced by the loss of KCNV1/Kv8.1 was rescued by knocking down Kv2.2, suggesting a potential Kv2.2-dependent downstream mechanism in cell death. Our study reveals, therefore, unsuspected and distinct roles of Kv8.1 and Kv2.2 in amyotrophic lateral sclerosis-related neurodegeneration.
Collapse
Affiliation(s)
- Xuan Huang
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Seungkyu Lee
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Kuchuan Chen
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Riki Kawaguchi
- Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ole Wiskow
- Department of Stem Cell and Regenerative Biology and Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Sulagna Ghosh
- Department of Stem Cell and Regenerative Biology and Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Devlin Frost
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Laura Perrault
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Roshan Pandey
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Joseph R Klim
- Department of Stem Cell and Regenerative Biology and Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Bruno Boivin
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Crystal Hermawan
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
| | - Kenneth J Livak
- Translational Immunogenomics Lab, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Daniel H Geschwind
- Department of Psychiatry and Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Brian J Wainger
- Department of Neurology, Mass General Brigham and Harvard Medical School, Boston, MA 02114, USA
| | - Kevin C Eggan
- Department of Stem Cell and Regenerative Biology and Department of Molecular and Cellular Biology, Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Bruce P Bean
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| | - Clifford J Woolf
- F.M. Kirby Neurobiology Research Center, Boston Children’s Hospital, Boston, MA 02115, USA
- Department of Neurobiology, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
61
|
Chen K, Forrest AM, Burgos GG, Kozai TDY. Neuronal functional connectivity is impaired in a layer dependent manner near chronically implanted intracortical microelectrodes in C57BL6 wildtype mice. J Neural Eng 2024; 21:036033. [PMID: 38788704 DOI: 10.1088/1741-2552/ad5049] [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: 11/06/2023] [Accepted: 05/24/2024] [Indexed: 05/26/2024]
Abstract
Objective.This study aims to reveal longitudinal changes in functional network connectivity within and across different brain structures near chronically implanted microelectrodes. While it is well established that the foreign-body response (FBR) contributes to the gradual decline of the signals recorded from brain implants over time, how the FBR affects the functional stability of neural circuits near implanted brain-computer interfaces (BCIs) remains unknown. This research aims to illuminate how the chronic FBR can alter local neural circuit function and the implications for BCI decoders.Approach.This study utilized single-shank, 16-channel,100µm site-spacing Michigan-style microelectrodes (3 mm length, 703µm2 site area) that span all cortical layers and the hippocampal CA1 region. Sex balanced C57BL6 wildtype mice (11-13 weeks old) received perpendicularly implanted microelectrode in left primary visual cortex. Electrophysiological recordings were performed during both spontaneous activity and visual sensory stimulation. Alterations in neuronal activity near the microelectrode were tested assessing cross-frequency synchronization of local field potential (LFP) and spike entrainment to LFP oscillatory activity throughout 16 weeks after microelectrode implantation.Main results. The study found that cortical layer 4, the input-receiving layer, maintained activity over the implantation time. However, layers 2/3 rapidly experienced severe impairment, leading to a loss of proper intralaminar connectivity in the downstream output layers 5/6. Furthermore, the impairment of interlaminar connectivity near the microelectrode was unidirectional, showing decreased connectivity from Layers 2/3 to Layers 5/6 but not the reverse direction. In the hippocampus, CA1 neurons gradually became unable to properly entrain to the surrounding LFP oscillations.Significance. This study provides a detailed characterization of network connectivity dysfunction over long-term microelectrode implantation periods. This new knowledge could contribute to the development of targeted therapeutic strategies aimed at improving the health of the tissue surrounding brain implants and potentially inform engineering of adaptive decoders as the FBR progresses. Our study's understanding of the dynamic changes in the functional network over time opens the door to developing interventions for improving the long-term stability and performance of intracortical microelectrodes.
Collapse
Affiliation(s)
- Keying Chen
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | - Adam M Forrest
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
| | | | - Takashi D Y Kozai
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, United States of America
- Center for Neural Basis of Cognition, Pittsburgh, PA, United States of America
- Center for Neuroscience, University of Pittsburgh, Pittsburgh, PA, United States of America
- McGowan Institute of Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, United States of America
- NeuroTech Center, University of Pittsburgh Brain Institute, Pittsburgh, PA, United States of America
| |
Collapse
|
62
|
Planas AM. Role of microglia in stroke. Glia 2024; 72:1016-1053. [PMID: 38173414 DOI: 10.1002/glia.24501] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Microglia play key roles in the post-ischemic inflammatory response and damaged tissue removal reacting rapidly to the disturbances caused by ischemia and working to restore the lost homeostasis. However, the modified environment, encompassing ionic imbalances, disruption of crucial neuron-microglia interactions, spreading depolarization, and generation of danger signals from necrotic neurons, induce morphological and phenotypic shifts in microglia. This leads them to adopt a proinflammatory profile and heighten their phagocytic activity. From day three post-ischemia, macrophages infiltrate the necrotic core while microglia amass at the periphery. Further, inflammation prompts a metabolic shift favoring glycolysis, the pentose-phosphate shunt, and lipid synthesis. These shifts, combined with phagocytic lipid intake, drive lipid droplet biogenesis, fuel anabolism, and enable microglia proliferation. Proliferating microglia release trophic factors contributing to protection and repair. However, some microglia accumulate lipids persistently and transform into dysfunctional and potentially harmful foam cells. Studies also showed microglia that either display impaired apoptotic cell clearance, or eliminate synapses, viable neurons, or endothelial cells. Yet, it will be essential to elucidate the viability of engulfed cells, the features of the local environment, the extent of tissue damage, and the temporal sequence. Ischemia provides a rich variety of region- and injury-dependent stimuli for microglia, evolving with time and generating distinct microglia phenotypes including those exhibiting proinflammatory or dysfunctional traits and others showing pro-repair features. Accurate profiling of microglia phenotypes, alongside with a more precise understanding of the associated post-ischemic tissue conditions, is a necessary step to serve as the potential foundation for focused interventions in human stroke.
Collapse
Affiliation(s)
- Anna M Planas
- Cerebrovascular Research Laboratory, Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Cerebrovascular Diseases, Area of Clinical and Experimental Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Barcelona, Spain
| |
Collapse
|
63
|
Mokbel AY, Burns MP, Main BS. The contribution of the meningeal immune interface to neuroinflammation in traumatic brain injury. J Neuroinflammation 2024; 21:135. [PMID: 38802931 PMCID: PMC11131220 DOI: 10.1186/s12974-024-03122-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 05/03/2024] [Indexed: 05/29/2024] Open
Abstract
Traumatic brain injury (TBI) is a major cause of disability and mortality worldwide, particularly among the elderly, yet our mechanistic understanding of what renders the post-traumatic brain vulnerable to poor outcomes, and susceptible to neurological disease, is incomplete. It is well established that dysregulated and sustained immune responses elicit negative consequences after TBI; however, our understanding of the neuroimmune interface that facilitates crosstalk between central and peripheral immune reservoirs is in its infancy. The meninges serve as the interface between the brain and the immune system, facilitating important bi-directional roles in both healthy and disease settings. It has been previously shown that disruption of this system exacerbates neuroinflammation in age-related neurodegenerative disorders such as Alzheimer's disease; however, we have an incomplete understanding of how the meningeal compartment influences immune responses after TBI. In this manuscript, we will offer a detailed overview of the holistic nature of neuroinflammatory responses in TBI, including hallmark features observed across clinical and animal models. We will highlight the structure and function of the meningeal lymphatic system, including its role in immuno-surveillance and immune responses within the meninges and the brain. We will provide a comprehensive update on our current knowledge of meningeal-derived responses across the spectrum of TBI, and identify new avenues for neuroimmune modulation within the neurotrauma field.
Collapse
Affiliation(s)
- Alaa Y Mokbel
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Mark P Burns
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA
| | - Bevan S Main
- Department of Neuroscience, Georgetown University Medical Center, New Research Building-EG11, 3970 Reservoir Rd, NW, Washington, DC, 20057, USA.
| |
Collapse
|
64
|
Pesti I, Légrádi Á, Farkas E. Primary microglia cell cultures in translational research: Strengths and limitations. J Biotechnol 2024; 386:10-18. [PMID: 38519034 DOI: 10.1016/j.jbiotec.2024.03.005] [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: 02/08/2024] [Revised: 03/13/2024] [Accepted: 03/13/2024] [Indexed: 03/24/2024]
Abstract
Microglia are the resident macrophages in the central nervous system, accounting for 10-15% of the cell mass in the brain. Next to their physiological role in development, monitoring neuronal function and the maintenance of homeostasis, microglia are crucial in the brain's immune defense. Brain injury and chronic neurological disorders are associated with neuroinflammation, in which microglia activation is a central element. Microglia acquire a wide spectrum of activation states in the diseased or injured brain, some of which are neurotoxic. The investigation of microglia (patho)physiology and therapeutic interventions targeting neuroinflammation is a substantial challenge. In addition to in vivo approaches, the application of in vitro model systems has gained significant ground and is essential to complement in vivo work. Primary microglia cultures have proved to be a useful tool. Microglia cultures have offered the opportunity to explore the mechanistic, molecular elements of microglia activation, the microglia secretome, and the efficacy of therapeutic treatments against neuroinflammation. As all model systems, primary microglia cultures have distinct strengths and limitations to be weighed when experiments are designed and when data are interpreted. Here, we set out to provide a succinct overview of the advantages and pitfalls of the use of microglia cultures, which instructs the refinement and further development of this technique to remain useful in the toolbox of microglia researchers. Since there is no conclusive therapy to combat neurotoxicity linked to neuroinflammation in acute brain injury or neurodegenerative disorders, these research tools remain essential to explore therapeutic opportunities.
Collapse
Affiliation(s)
- István Pesti
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Somogyi u 4, Szeged 6720, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Somogyi u 4, Szeged 6720, Hungary
| | - Ádám Légrádi
- Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Somogyi u 4, Szeged 6720, Hungary
| | - Eszter Farkas
- Hungarian Centre of Excellence for Molecular Medicine - University of Szeged Cerebral Blood Flow and Metabolism Research Group, Somogyi u 4, Szeged 6720, Hungary; Department of Cell Biology and Molecular Medicine, Albert Szent-Györgyi Medical School and Faculty of Science and Informatics, University of Szeged, Somogyi u 4, Szeged 6720, Hungary.
| |
Collapse
|
65
|
Lénárt N, Cserép C, Császár E, Pósfai B, Dénes Á. Microglia-neuron-vascular interactions in ischemia. Glia 2024; 72:833-856. [PMID: 37964690 DOI: 10.1002/glia.24487] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 10/24/2023] [Accepted: 10/24/2023] [Indexed: 11/16/2023]
Abstract
Cerebral ischemia is a devastating condition that results in impaired blood flow in the brain leading to acute brain injury. As the most common form of stroke, occlusion of cerebral arteries leads to a characteristic sequence of pathophysiological changes in the brain tissue. The mechanisms involved, and comorbidities that determine outcome after an ischemic event appear to be highly heterogeneous. On their own, the processes leading to neuronal injury in the absence of sufficient blood supply to meet the metabolic demand of the cells are complex and manifest at different temporal and spatial scales. While the contribution of non-neuronal cells to stroke pathophysiology is increasingly recognized, recent data show that microglia, the main immune cells of the central nervous system parenchyma, play previously unrecognized roles in basic physiological processes beyond their inflammatory functions, which markedly change during ischemic conditions. In this review, we aim to discuss some of the known microglia-neuron-vascular interactions assumed to contribute to the acute and delayed pathologies after cerebral ischemia. Because the mechanisms of neuronal injury have been extensively discussed in several excellent previous reviews, here we focus on some recently explored pathways that may directly or indirectly shape neuronal injury through microglia-related actions. These discoveries suggest that modulating gliovascular processes in different forms of stroke and other neurological disorders might have presently unexplored therapeutic potential in combination with neuroprotective and flow restoration strategies.
Collapse
Affiliation(s)
- Nikolett Lénárt
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Csaba Cserép
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Császár
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- Momentum Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| |
Collapse
|
66
|
Surala M, Soso-Zdravkovic L, Munro D, Rifat A, Ouk K, Vida I, Priller J, Madry C. Lifelong absence of microglia alters hippocampal glutamatergic networks but not synapse and spine density. EMBO Rep 2024; 25:2348-2374. [PMID: 38589666 PMCID: PMC11094096 DOI: 10.1038/s44319-024-00130-9] [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: 02/27/2024] [Revised: 03/13/2024] [Accepted: 03/20/2024] [Indexed: 04/10/2024] Open
Abstract
Microglia sculpt developing neural circuits by eliminating excess synapses in a process called synaptic pruning, by removing apoptotic neurons, and by promoting neuronal survival. To elucidate the role of microglia during embryonic and postnatal brain development, we used a mouse model deficient in microglia throughout life by deletion of the fms-intronic regulatory element (FIRE) in the Csf1r locus. Surprisingly, young adult Csf1rΔFIRE/ΔFIRE mice display no changes in excitatory and inhibitory synapse number and spine density of CA1 hippocampal neurons compared with Csf1r+/+ littermates. However, CA1 neurons are less excitable, receive less CA3 excitatory input and show altered synaptic properties, but this does not affect novel object recognition. Cytokine profiling indicates an anti-inflammatory state along with increases in ApoE levels and reactive astrocytes containing synaptic markers in Csf1rΔFIRE/ΔFIRE mice. Notably, these changes in Csf1rΔFIRE/ΔFIRE mice closely resemble the effects of acute microglial depletion in adult mice after normal development. Our findings suggest that microglia are not mandatory for synaptic pruning, and that in their absence pruning can be achieved by other mechanisms.
Collapse
Affiliation(s)
- Michael Surala
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
| | - Luna Soso-Zdravkovic
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
| | - David Munro
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, EH16 4TJ, UK
| | - Ali Rifat
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Koliane Ouk
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charitéplatz 1, 10117, Berlin, Germany
| | - Imre Vida
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute for Integrative Neuroanatomy, Charitéplatz 1, 10117, Berlin, Germany
| | - Josef Priller
- University of Edinburgh and UK Dementia Research Institute, Edinburgh, EH16 4TJ, UK.
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Neuropsychiatry and Laboratory of Molecular Psychiatry, Charitéplatz 1, 10117, Berlin, Germany.
- DZNE Berlin, 10117, Berlin, Germany.
- Department of Psychiatry and Psychotherapy; School of Medicine and Health, Technical University of Munich and German Center for Mental Health (DZPG), 81675, Munich, Germany.
| | - Christian Madry
- Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Institute of Neurophysiology, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
67
|
Escoubas CC, Molofsky AV. Microglia as integrators of brain-associated molecular patterns. Trends Immunol 2024; 45:358-370. [PMID: 38658221 DOI: 10.1016/j.it.2024.03.009] [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: 03/01/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/26/2024]
Abstract
Microglia are brain-resident macrophages that play key roles in brain development and experience dependent plasticity. In this review we discuss recent findings regarding the molecular mechanisms through which mammalian microglia sense the unique molecular patterns of the homeostatic brain. We propose that microglial function is acutely controlled in response to 'brain-associated molecular patterns' (BAMPs) that function as indicators of neuronal activity and neural circuit remodeling. A further layer of regulation comes from instructive cytokine cues that define unique microglial functional states. A systematic investigation of the receptors and signaling pathways that mediate these two regulatory axes may begin to define a functional code for microglia-neuron interactions.
Collapse
Affiliation(s)
- Caroline C Escoubas
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA
| | - Anna V Molofsky
- Departments of Psychiatry and Behavioral Sciences/Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94158, USA; Kavli Institute for Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94158, USA; Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
68
|
Wang P, Mi Y, Yu H, Teng X, Jin S, Xiao L, Xue H, Tian D, Guo Q, Wu Y. Trimethylamine-N-oxide aggravated the sympathetic excitation in D-galactose induced aging rats by down-regulating P2Y12 receptor in microglia. Biomed Pharmacother 2024; 174:116549. [PMID: 38593701 DOI: 10.1016/j.biopha.2024.116549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 03/26/2024] [Accepted: 04/04/2024] [Indexed: 04/11/2024] Open
Abstract
This study aimed to determine whether trimethylamine N-oxide (TMAO) was involved in sympathetic activation in aging and the underlying mechanisms. Our hypothesis is TMAO reduces P2Y12 receptor (P2Y12R) and induces microglia-mediated inflammation in the paraventricular nucleus (PVN), then leading to sympathetic activation in aging. This study involved 18 young adults and 16 old adults. Aging rats were established by injecting D-galactose (D-gal, 200 mg/kg/d) subcutaneously for 12 weeks. TMAO (120 mg/kg/d) or 1% 3, 3-dimethyl-l-butanol (DMB) was administrated via drinking water for 12 weeks to investigate their effects on neuroinflammation and sympathetic activation in aging rats. Plasma TMAO, NE and IL-1β levels were higher in old adults than in young adults. In addition, standard deviation of all normal to normal intervals (SDNN) and standard deviation of the average of normal to normal intervals (SDANN) were lower in old adults and negatively correlated with TMAO, indicating sympathetic activation in old adults, which is associated with an increase in TMAO levels. Treatment of rats with D-gal showed increased senescence-associated protein levels and microglia-mediated inflammation, as well as decreased P2Y12R protein levels in PVN. Plasma TMAO, NE and IL-1β levels were increased, accompanied by enhanced renal sympathetic nerve activity (RSNA). While TMAO treatment exacerbated the above phenomenon, DMB mitigated it. These findings suggest that TMAO contributes to sympathetic hyperactivity in aging by downregulating P2Y12R in microglia and increasing inflammation in the PVN. These results may provide promising new target for the prevention and treatment of aging and aging-related diseases.
Collapse
Affiliation(s)
- Ping Wang
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Yuan Mi
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Department of Emergency, the Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, China
| | - Hao Yu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Xu Teng
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Sheng Jin
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Lin Xiao
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Hongmei Xue
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Danyang Tian
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China
| | - Qi Guo
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Experimental Center for Teaching, Hebei Medical University, Shijiazhuang 050017, China; Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, China.
| | - Yuming Wu
- Department of Physiology, Institute of Basic Medicine, Hebei Medical University, 361 Zhongshan East Road, Shijiazhuang 050017, China; Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Shijiazhuang 050017, China; The Key Laboratory of Neural and Vascular Biology, Ministry of Education, China; Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, China.
| |
Collapse
|
69
|
Wang Y, Song Y, Zhang L, Huang X. The paradoxical role of zinc on microglia. J Trace Elem Med Biol 2024; 83:127380. [PMID: 38171037 DOI: 10.1016/j.jtemb.2023.127380] [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: 09/26/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/05/2024]
Abstract
Zinc is an essential trace element for humans, and its homeostasis is essential for the health of the central nervous system. Microglia, the resident immune cells in the central nervous system, play the roles of sustaining, nourishing, and immune surveillance. Microglia are sensitive to microenvironment changes and are easily activated to M1 phenotype to enhance disease progression or the M2 phenotype to improve peripheral nerves injury repair. Zinc is requisite for microglial activation, However, the cytotoxicity outcome of zinc against microglia, the activated microglia phenotype, and activated microglia function are ambiguous. Herein, we have reviewed the neurological function of zinc and microglia, particularly the ambiguous role of zinc on microglia. We also pay attention to the role of zinc homeostasis on microglial function within the central nervous system disease. Finally, we observe the relationship between zinc and microglia, attempting to design new therapeutic measures against major nervous system disorders.
Collapse
Affiliation(s)
- Yehong Wang
- Graduate Faculty, Xi'an Physical Education University, Xi'an 710068, PR China; Hunan Provincial Key Laboratory of Dong Medicine, Ethnic Medicine Research Center, Hunan University of Medicine, Huaihua 418000, PR China
| | - Yi Song
- Department of Neurosurgery, Chongqing University Three Gorges Hospital, Chongqing 404100, PR China.
| | - Lingdang Zhang
- Department of Neurosurgery, Chongqing University Three Gorges Hospital, Chongqing 404100, PR China
| | - Xiao Huang
- Hunan Provincial Key Laboratory of Dong Medicine, Ethnic Medicine Research Center, Hunan University of Medicine, Huaihua 418000, PR China.
| |
Collapse
|
70
|
Zhang L, Liu J, Liu M. Transsynaptic degeneration of ventral horn motor neurons exists but plays a minor role in lower motor system dysfunction in acute ischemic rats. PLoS One 2024; 19:e0298006. [PMID: 38669239 PMCID: PMC11051614 DOI: 10.1371/journal.pone.0298006] [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: 08/30/2023] [Accepted: 01/16/2024] [Indexed: 04/28/2024] Open
Abstract
BACKGROUND As a leading cause of mortality and long-term disability, acute ischemic stroke can produce far-reaching pathophysiological consequences. Accumulating evidence has demonstrated abnormalities in the lower motor system following stroke, while the existence of Transsynaptic degeneration of contralateral spinal cord ventral horn (VH) neurons is still debated. METHODS Using a rat model of acute ischemic stroke, we analyzed spinal cord VH neuron counts contralaterally and ipsilaterally after stroke with immunofluorescence staining. Furthermore, we estimated the overall lower motor unit abnormalities after stroke by simultaneously measuring the modified neurological severity score (mNSS), compound muscle action potential (CMAP) amplitude, repetitive nerve stimulation (RNS), spinal cord VH neuron counts, and the corresponding muscle fiber morphology. The activation status of microglia and extracellular signal-regulated kinase 1/2 (ERK 1/2) in the spinal cord VH was also assessed. RESULTS At 7 days after stroke, the contralateral CMAP amplitudes declined to a nadir indicating lower motor function damage, and significant muscle disuse atrophy was observed on the same side; meanwhile, the VH neurons remained intact. At 14 days after focal stroke, lower motor function recovered with alleviated muscle disuse atrophy, while transsynaptic degeneration occurred on the contralateral side with elevated activation of ERK 1/2, along with the occurrence of neurogenic muscle atrophy. No apparent decrement of CMAP amplitude was observed with RNS during the whole experimental process. CONCLUSIONS This study offered an overview of changes in the lower motor system in experimental ischemic rats. We demonstrated that transsynaptic degeneration of contralateral VH neurons occurred when lower motor function significantly recovered, which indicated the minor role of transsynaptic degeneration in lower motor dysfunction during the acute and subacute phases of focal ischemic stroke.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Jingwen Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingsheng Liu
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
71
|
Chinnathambi S, Chidambaram H. G-protein coupled receptors regulates Tauopathy in neurodegeneration. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:467-493. [PMID: 38960483 DOI: 10.1016/bs.apcsb.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
In Alzheimer's disease, the microtubule-associated protein, Tau misfolds to form aggregates and filaments in the intra- and extracellular region of neuronal cells. Microglial cells are the resident brain macrophage cells involved in constant surveillance and activated by the extracellular deposits. Purinergic receptors are involved in the chemotactic migration of microglial cells towards the site of inflammation. From our recent study, we have observed that the microglial P2Y12 receptor is involved in phagocytosis of full-length Tau species such as monomers, oligomers and aggregates by actin-driven chemotaxis. This study shows the interaction of repeat-domain of Tau (TauRD) with the microglial P2Y12 receptor and the corresponding residues for interaction have been analyzed by various in-silico approaches. In the cellular studies, TauRD was found to interact with microglial P2Y12R and induces its cellular expression confirmed by co-immunoprecipitation and western blot analysis. Furthermore, the P2Y12R-mediated TauRD internalization has demonstrated activation of microglia with an increase in the Iba1 level, and TauRD becomes accumulated at the peri-nuclear region for the degradation.
Collapse
Affiliation(s)
- Subashchandrabose Chinnathambi
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India.
| | - Hariharakrishnan Chidambaram
- Department of Neurochemistry, National Institute of Mental Health and Neuro Sciences (NIMHANS), Institute of National Importance, Bangalore, Karnataka, India
| |
Collapse
|
72
|
Yang Q, Vazquez AL, Cui XT. Revealing in vivo cellular mechanisms of cerebral microbleeds on neurons and microglia across cortical layers. iScience 2024; 27:109371. [PMID: 38510113 PMCID: PMC10951986 DOI: 10.1016/j.isci.2024.109371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 12/28/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Cerebral microbleeds (CMBs) are associated with higher risk for various neurological diseases including stroke, dementia, and Alzheimer's disease. However, the understanding of cellular pathology of CMBs, particularly in deep brain regions, remains limited. Utilizing two-photon microscopy and microprism implantation, we longitudinally imaged the impact of CMBs on neuronal and microglial activities across cortical depths in awake mice. A temporary decline in spontaneous neuronal activity occurred throughout cortical layers, followed by recovery within a week. However, significant changes of neuron-neuron activity correlations persisted for weeks. Moreover, microglial contact with neuron soma significantly increased post-microbleeds, indicating an important modulatory role of microglia. Notably, microglial contact, negatively correlated with neuronal firing rate in normal conditions, became uncorrelated after microbleeds, suggesting a decreased neuron-microglia inhibition. These findings reveal chronic alterations in cortical neuronal networks and microglial-neuronal interactions across cortical depths, shedding light on the pathology of CMBs.
Collapse
Affiliation(s)
- Qianru Yang
- Department of Neurosurgery, Stanford University, Palo Alto, CA, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
| | - Alberto L. Vazquez
- Center for Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
- Department of Radiology, University of Pittsburgh, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - X. Tracy Cui
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
- Center for Neural Basis of Cognition, University of Pittsburgh and Carnegie Mellon University, Pittsburgh, PA, USA
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
73
|
Song N, Mei S, Wang X, Hu G, Lu M. Focusing on mitochondria in the brain: from biology to therapeutics. Transl Neurodegener 2024; 13:23. [PMID: 38632601 PMCID: PMC11022390 DOI: 10.1186/s40035-024-00409-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 03/13/2024] [Indexed: 04/19/2024] Open
Abstract
Mitochondria have multiple functions such as supplying energy, regulating the redox status, and producing proteins encoded by an independent genome. They are closely related to the physiology and pathology of many organs and tissues, among which the brain is particularly prominent. The brain demands 20% of the resting metabolic rate and holds highly active mitochondrial activities. Considerable research shows that mitochondria are closely related to brain function, while mitochondrial defects induce or exacerbate pathology in the brain. In this review, we provide comprehensive research advances of mitochondrial biology involved in brain functions, as well as the mitochondria-dependent cellular events in brain physiology and pathology. Furthermore, various perspectives are explored to better identify the mitochondrial roles in neurological diseases and the neurophenotypes of mitochondrial diseases. Finally, mitochondrial therapies are discussed. Mitochondrial-targeting therapeutics are showing great potentials in the treatment of brain diseases.
Collapse
Affiliation(s)
- Nanshan Song
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Shuyuan Mei
- The First Clinical Medical College, Nanjing Medical University, Nanjing, 211166, China
| | - Xiangxu Wang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China
| | - Gang Hu
- Department of Pharmacology, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Neuroprotective Drug Discovery Key Laboratory, School of Basic Medical Sciences, Nanjing Medical University, Nanjing, 211166, China.
- Changzhou Second People's Hospital, Changzhou Medical Center, Nanjing Medical University, Changzhou, 213000, China.
| |
Collapse
|
74
|
Uweru OJ, Okojie AK, Trivedi A, Benderoth J, Thomas LS, Davidson G, Cox K, Eyo UB. A P2RY12 deficiency results in sex-specific cellular perturbations and sexually dimorphic behavioral anomalies. J Neuroinflammation 2024; 21:95. [PMID: 38622726 PMCID: PMC11017545 DOI: 10.1186/s12974-024-03079-7] [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: 12/18/2023] [Accepted: 03/28/2024] [Indexed: 04/17/2024] Open
Abstract
Microglia are sexually dimorphic, yet, this critical aspect is often overlooked in neuroscientific studies. Decades of research have revealed the dynamic nature of microglial-neuronal interactions, but seldom consider how this dynamism varies with microglial sex differences, leaving a significant gap in our knowledge. This study focuses on P2RY12, a highly expressed microglial signature gene that mediates microglial-neuronal interactions, we show that adult females have a significantly higher expression of the receptor than adult male microglia. We further demonstrate that a genetic deletion of P2RY12 induces sex-specific cellular perturbations with microglia and neurons in females more significantly affected. Correspondingly, female mice lacking P2RY12 exhibit unique behavioral anomalies not observed in male counterparts. These findings underscore the critical, sex-specific roles of P2RY12 in microglial-neuronal interactions, offering new insights into basal interactions and potential implications for CNS disease mechanisms.
Collapse
Affiliation(s)
- Ogochukwu J Uweru
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA.
| | - Akhabue K Okojie
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Aparna Trivedi
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
| | - Jordan Benderoth
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Lauren S Thomas
- North Carolina Agricultural and Technical State University, Greensboro, NC, USA
| | - Georgia Davidson
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Kendall Cox
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B Eyo
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA.
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA.
- Neuroscience Graduate Program, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
75
|
Kim W, Kim M, Kim B. Unraveling the enigma: housekeeping gene Ugt1a7c as a universal biomarker for microglia. Front Psychiatry 2024; 15:1364201. [PMID: 38666091 PMCID: PMC11043603 DOI: 10.3389/fpsyt.2024.1364201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 03/26/2024] [Indexed: 04/28/2024] Open
Abstract
Background Microglia, brain resident macrophages, play multiple roles in maintaining homeostasis, including immunity, surveillance, and protecting the central nervous system through their distinct activation processes. Identifying all types of microglia-driven populations is crucial due to the presence of various phenotypes that differ based on developmental stages or activation states. During embryonic development, the E8.5 yolk sac contains erythromyeloid progenitors that go through different growth phases, eventually resulting in the formation of microglia. In addition, microglia are present in neurological diseases as a diverse population. So far, no individual biomarker for microglia has been discovered that can accurately identify and monitor their development and attributes. Summary Here, we highlight the newly defined biomarker of mouse microglia, UGT1A7C, which exhibits superior stability in expression during microglia development and activation compared to other known microglia biomarkers. The UGT1A7C sensing chemical probe labels all microglia in the 3xTG AD mouse model. The expression of Ugt1a7c is stable during development, with only a 4-fold variation, while other microglia biomarkers, such as Csf1r and Cx3cr1, exhibit at least a 10-fold difference. The UGT1A7C expression remains constant throughout its lifespan. In addition, the expression and activity of UGT1A7C are the same in response to different types of inflammatory activators' treatment in vitro. Conclusion We propose employing UGT1A7C as the representative biomarker for microglia, irrespective of their developmental state, age, or activation status. Using UGT1A7C can reduce the requirement for using multiple biomarkers, enhance the precision of microglia analysis, and even be utilized as a standard for gene/protein expression.
Collapse
Affiliation(s)
| | | | - Beomsue Kim
- Neural Circuit Research Group, Korea Brain Research Institute, Daegu, Republic of Korea
| |
Collapse
|
76
|
Binder LB, Rosa PB, de Sousa BM, Chagas LS, Dubljević O, Martineau FS, Mottarlini F, Castany S, Morton L, Krstanović F, Tassinari ID, Choconta JL, Pereira-Santos AR, Weinhard L, Pallegar PN, Vahsen BF, Lepiarz-Raba I, Compagnion AC, Lorente-Picón M. Neuro-immune interactions in health and disease: Insights from FENS-Hertie 2022 Winter School. Eur J Neurosci 2024; 59:1977-1992. [PMID: 38311960 DOI: 10.1111/ejn.16262] [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: 10/04/2023] [Revised: 12/16/2023] [Accepted: 01/07/2024] [Indexed: 02/06/2024]
Abstract
In a great partnership, the Federation of European Neuroscience Societies (FENS) and the Hertie Foundation organized the FENS-Hertie 2022 Winter School on 'Neuro-immune interactions in health and disease'. The school selected 27 PhD students and 13 postdoctoral fellows from 20 countries and involved 14 faculty members experts in the field. The Winter School focused on a rising field of research, the interactions between the nervous and both innate and adaptive immune systems under pathological and physiological conditions. A fine-tuned neuro-immune crosstalk is fundamental for healthy development, while disrupted neuro-immune communication might play a role in neurodegeneration, neuroinflammation and aging. However, much is yet to be understood about the underlying mechanisms of these neuro-immune interactions in the healthy brain and under pathological scenarios. In addition to new findings in this emerging field, novel methodologies and animal models were presented to foment research on neuro-immunology. The FENS-Hertie 2022 Winter School provided an insightful knowledge exchange between students and faculty focusing on the latest discoveries in the biology of neuro-immune interactions while fostering great academic and professional opportunities for early-career neuroscientists from around the world.
Collapse
Affiliation(s)
- Luisa B Binder
- Department of Psychiatry and Neuroscience, Faculty of Medicine and CERVO Brain Research Center, Université Laval, Quebec City, Canada
| | - Priscila B Rosa
- Center for Social and Affective Neuroscience (CSAN), Linköping University, Linköping, Sweden
| | - Bárbara M de Sousa
- Institute of Biomedicine (iBiMED), Department of Medical Sciences, Universidade de Aveiro, Aveiro, Portugal
| | - Luana S Chagas
- Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Olga Dubljević
- Department of Neurobiology, Univerzitet u Beogradu Institut za Biološka Istraživanja Siniša Stanković, Institute for Biological Research, Beograd, Republic of Serbia
| | | | - Francesca Mottarlini
- Department of Pharmacological and Biomolecular Sciences 'Rodolfo Paoletti', Università degli Studi di Milano, Milan, Italy
| | - Sílvia Castany
- Center for Social and Affective Neuroscience (CSAN), Linköping University, Linköping, Sweden
| | - Lorena Morton
- Faculty of Medicine, Institute of Inflammation and Neurodegeneration, Otto-von-Guericke University, Magdeburg, Germany
| | - Fran Krstanović
- Faculty of Medicine, Center for Proteomics, University of Rijeka, Rijeka, Croatia
| | - Isadora D Tassinari
- Department of Physiology, Graduate Program in Physiology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Jeiny L Choconta
- Institute of Physiology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ana Raquel Pereira-Santos
- Center for Neuroscience and Cell Biology (CNC), CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
| | | | | | - Björn F Vahsen
- Nuffield Department of Clinical Neurosciences, Oxford Motor Neuron Disease Centre, University of Oxford, John Radcliffe Hospital, Oxford, UK
- Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, UK
| | - Izabela Lepiarz-Raba
- BRAINCITY: Centre of Excellence for Neural Plasticity and Brain Disorders, Nencki Institute of Experimental Biology PAS, Warsaw, Poland
| | | | - Marina Lorente-Picón
- Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| |
Collapse
|
77
|
Guo Y, Mao T, Fang Y, Wang H, Yu J, Zhu Y, Shen S, Zhou M, Li H, Hu Q. Comprehensive insights into potential roles of purinergic P2 receptors on diseases: Signaling pathways involved and potential therapeutics. J Adv Res 2024:S2090-1232(24)00123-1. [PMID: 38565403 DOI: 10.1016/j.jare.2024.03.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/03/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Purinergic P2 receptors, which can be divided into ionotropic P2X receptors and metabotropic P2Y receptors, mediate cellular signal transduction of purine or pyrimidine nucleoside triphosphates and diphosphate. Based on the wide expression of purinergic P2 receptors in tissues and organs, their significance in homeostatic maintenance, metabolism, nociceptive transmission, and other physiological processes is becoming increasingly evident, suggesting that targeting purinergic P2 receptors to regulate biological functions and signal transmission holds significant promise for disease treatment. AIM OF REVIEW This review highlights the detailed mechanisms by which purinergic P2 receptors engage in physiological and pathological progress, as well as providing prospective strategies for discovering clinical drug candidates. KEY SCIENTIFIC CONCEPTS OF REVIEW The purinergic P2 receptors regulate complex signaling and molecular mechanisms in nervous system, digestive system, immune system and as a result, controlling physical health states and disease progression. There has been a significant rise in research and development focused on purinergic P2 receptors, contributing to an increased number of drug candidates in clinical trials. A few influential pioneers have laid the foundation for advancements in the evaluation, development, and of novel purinergic P2 receptors modulators, including agonists, antagonists, pharmaceutical compositions and combination strategies, despite the different scaffolds of these drug candidates. These advancements hold great potential for improving therapeutic outcomes by specifically targeting purinergic P2 receptors.
Collapse
Affiliation(s)
- Yanshuo Guo
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Tianqi Mao
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Yafei Fang
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hui Wang
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Jiayue Yu
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yifan Zhu
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China
| | - Shige Shen
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Mengze Zhou
- School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China.
| | - Huanqiu Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou 215006, China.
| | - Qinghua Hu
- School of Life Science and Technology, China Pharmaceutical University, Nanjing 211198, China.
| |
Collapse
|
78
|
So WY, Johnson B, Gordon PB, Bishop KS, Gong H, Burr HA, Staunton JR, Handler C, Sood R, Scarcelli G, Tanner K. Macrophage mediated mesoscale brain mechanical homeostasis mechanically imaged via optical tweezers and Brillouin microscopy in vivo. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.12.27.573380. [PMID: 38234798 PMCID: PMC10793422 DOI: 10.1101/2023.12.27.573380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2024]
Abstract
Tissues are active materials where epithelial turnover, immune surveillance, and remodeling of stromal cells such as macrophages all regulate form and function. Scattering modalities such as Brillouin microscopy (BM) can non-invasively access mechanical signatures at GHz. However, our traditional understanding of tissue material properties is derived mainly from modalities which probe mechanical properties at different frequencies. Thus, reconciling measurements amongst these modalities remains an active area. Here, we compare optical tweezer active microrheology (OT-AMR) and Brillouin microscopy (BM) to longitudinally map brain development in the larval zebrafish. We determine that each measurement is able to detect a mechanical signature linked to functional units of the brain. We demonstrate that the corrected BM-Longitudinal modulus using a density factor correlates well with OT-AMR storage modulus at lower frequencies. We also show that the brain tissue mechanical properties are dependent on both the neuronal architecture and the presence of macrophages. Moreover, the BM technique is able to delineate the contributions to mechanical properties of the macrophage from that due to colony stimulating factor 1 receptor (CSF1R) mediated stromal remodeling. Here, our data suggest that macrophage remodeling is instrumental in the maintenance of tissue mechanical homeostasis during development. Moreover, the strong agreement between the OT-AM and BM further demonstrates that scattering-based technique is sensitive to both large and minute structural modification in vivo.
Collapse
Affiliation(s)
- Woong Young So
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Bailey Johnson
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | | | - Kevin S. Bishop
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | - Hyeyeon Gong
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
- University of Maryland - College Park, MD, USA
| | - Hannah A Burr
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| | | | | | - Raman Sood
- National Human Genome Research Institute, NIH, MD, USA
| | | | - Kandice Tanner
- National Cancer Institute, National Institutes of Health (NIH), MD, USA
| |
Collapse
|
79
|
Hartmann SM, Heider J, Wüst R, Fallgatter AJ, Volkmer H. Microglia-neuron interactions in schizophrenia. Front Cell Neurosci 2024; 18:1345349. [PMID: 38510107 PMCID: PMC10950997 DOI: 10.3389/fncel.2024.1345349] [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: 11/27/2023] [Accepted: 02/21/2024] [Indexed: 03/22/2024] Open
Abstract
Multiple lines of evidence implicate increased neuroinflammation mediated by glial cells to play a key role in neurodevelopmental disorders such as schizophrenia. Microglia, which are the primary innate immune cells of the brain, are crucial for the refinement of the synaptic circuitry during early brain development by synaptic pruning and the regulation of synaptic plasticity during adulthood. Schizophrenia risk factors as genetics or environmental influences may further be linked to increased activation of microglia, an increase of pro-inflammatory cytokine levels and activation of the inflammasome resulting in an overall elevated neuroinflammatory state in patients. Synaptic loss, one of the central pathological hallmarks of schizophrenia, is believed to be due to excess removal of synapses by activated microglia, primarily affecting glutamatergic neurons. Therefore, it is crucial to investigate microglia-neuron interactions, which has been done by multiple studies focusing on post-mortem brain tissues, brain imaging, animal models and patient iPSC-derived 2D culture systems. In this review, we summarize the major findings in patients and in vivo and in vitro models in the context of neuron-microglia interactions in schizophrenia and secondly discuss the potential of anti-inflammatory treatments for the alleviation of positive, negative, and cognitive symptoms.
Collapse
Affiliation(s)
- Sophia-Marie Hartmann
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Johanna Heider
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| | - Richard Wüst
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Andreas J. Fallgatter
- Department of Psychiatry, Tübingen Center for Mental Health (TüCMH), University of Tübingen, Tübingen, Germany
| | - Hansjürgen Volkmer
- Molecular Neurobiology, Department of Pharma and Biotech, NMI Natural and Medical Sciences Institute at the University of Tübingen, Reutlingen, Germany
| |
Collapse
|
80
|
Durán Laforet V, Schafer DP. Microglia: Activity-dependent regulators of neural circuits. Ann N Y Acad Sci 2024; 1533:38-50. [PMID: 38294960 PMCID: PMC10976428 DOI: 10.1111/nyas.15105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2024]
Abstract
It has been more than a century since Pío del Río-Hortega first characterized microglia in histological stains of brain tissue. Since then, significant advances have been made in understanding the role of these resident central nervous system (CNS) macrophages. In particular, it is now known that microglia can sense neural activity and modulate neuronal circuits accordingly. We review the mechanisms by which microglia detect changes in neural activity to then modulate synapse numbers in the developing and mature CNS. This includes responses to both spontaneous and experience-driven neural activity. We further discuss activity-dependent mechanisms by which microglia regulate synaptic function and neural circuit excitability. Together, our discussion provides a comprehensive review of the activity-dependent functions of microglia within neural circuits in the healthy CNS, and highlights exciting new open questions related to understanding more fully microglia as key components and regulators of neural circuits.
Collapse
Affiliation(s)
- Violeta Durán Laforet
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Dorothy P Schafer
- Department of Neurobiology, Brudnick Neuropsychiatric Research Institute, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
81
|
Zhao S, Umpierre AD, Wu LJ. Tuning neural circuits and behaviors by microglia in the adult brain. Trends Neurosci 2024; 47:181-194. [PMID: 38245380 PMCID: PMC10939815 DOI: 10.1016/j.tins.2023.12.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 11/04/2023] [Accepted: 12/21/2023] [Indexed: 01/22/2024]
Abstract
Microglia are the primary immune cells of the CNS, contributing to both inflammatory damage and tissue repair in neurological disorder. In addition, emerging evidence highlights the role of homeostatic microglia in regulating neuronal activity, interacting with synapses, tuning neural circuits, and modulating behaviors. Herein, we review how microglia sense and regulate neuronal activity through synaptic interactions, thereby directly engaging with neural networks and behaviors. We discuss current studies utilizing microglial optogenetic and chemogenetic approaches to modulate adult neural circuits. These manipulations of microglia across different CNS regions lead to diverse behavioral consequences. We propose that spatial heterogeneity of microglia-neuron interaction lays the groundwork for understanding diverse functions of microglia in neural circuits and behaviors.
Collapse
Affiliation(s)
- Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Mayo Clinic Graduate School of Biomedical Sciences, Rochester, MN, USA
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA; Department of Immunology, Mayo Clinic, Rochester, MN, USA; Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
82
|
Kou L, Chi X, Sun Y, Yin S, Wu J, Zou W, Wang Y, Jin Z, Huang J, Xiong N, Xia Y, Wang T. Circadian regulation of microglia function: Potential targets for treatment of Parkinson's Disease. Ageing Res Rev 2024; 95:102232. [PMID: 38364915 DOI: 10.1016/j.arr.2024.102232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Revised: 02/11/2024] [Accepted: 02/11/2024] [Indexed: 02/18/2024]
Abstract
Circadian rhythms are involved in the regulation of many aspects of the body, including cell function, physical activity and disease. Circadian disturbance often predates the typical symptoms of neurodegenerative diseases and is not only a non-motor symptom, but also one of the causes of their occurrence and progression. Glial cells possess circadian clocks that regulate their function to maintain brain development and homeostasis. Emerging evidence suggests that the microglial circadian clock is involved in the regulation of many physiological processes, such as cytokine release, phagocytosis, and nutritional and metabolic support, and that disruption of the microglia clock may affect multiple aspects of Parkinson's disease, especially neuroinflammation and α-synuclein processes. Herein, we review recent advances in the circadian control of microglia function in health and disease, and discuss novel pharmacological interventions for microglial clocks in neurodegenerative disorders.
Collapse
Affiliation(s)
- Liang Kou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaosa Chi
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yadi Sun
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Sijia Yin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiawei Wu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wenkai Zou
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yiming Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zongjie Jin
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yun Xia
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
83
|
Zou D, Huang X, Lan Y, Pan M, Xie J, Huang Q, Zeng J, Zou C, Pei Z, Zou C, Mao Y, Luo J. Single-cell and spatial transcriptomics reveals that PTPRG activates the m 6A methyltransferase VIRMA to block mitophagy-mediated neuronal death in Alzheimer's disease. Pharmacol Res 2024; 201:107098. [PMID: 38325728 DOI: 10.1016/j.phrs.2024.107098] [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: 11/10/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 02/09/2024]
Abstract
Neuronal death is one of the key pathologies in Alzheimer's disease (AD). How neuronal death begins in AD is far from clear, so clarifying this process may help develop effective therapies. This study collected single-cell RNA sequencing data of 85 AD samples and 83 control samples, covering the prefrontal cortex, internal olfactory cortex, superior parietal lobe, superior frontal gyrus, caudal internal olfactory cortex, somatosensory cortex, hippocampus, superior frontal cortex and peripheral blood mononuclear cells. Additionally, spatial transcriptomic data of coronal sections from 6 AppNL-G-F AD mice and 6 control C57Bl/6 J mice were acquired. The main single-cell and spatial transcriptomics results were experimentally validated in wild type and 5 × FAD mice. We found that the microglia subpopulation Mic_PTPRG can communicate with specific types of neurons (especially excitatory ExNeu_PRKN_VIRMA and inhibitory InNeu_PRKN_VIRMA neuronal subpopulations) and cause them to express PTPRG during AD progression. Within neurons, PTPRG binds and upregulates the m6A methyltransferase VIRMA, thus inhibiting translation of PRKN mRNA to prevent the clearance of damaged mitochondria in neurons through suppressing mitophagy. As the disease progresses, the energy and nutrient metabolic pathways in neurons are reprogrammed, leading to their death. Consistently, we determined that PTPTRG can physically interact with VIRMA in mouse brains and PRKN is significantly upregulated in 5 × FAD mouse brain. Altogether, our findings demonstrate that PTPRG activates the m6A methyltransferase VIRMA to block mitophagy-mediated neuronal death in AD, which is a potential pathway, through which microglia and neuronal PTPRG modify neuronal connections in the brain during AD progression.
Collapse
Affiliation(s)
- Donghua Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China.
| | - Xiaohua Huang
- Department of Neurology, The Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, China
| | - Yating Lan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Mika Pan
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Jieqiong Xie
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Qi Huang
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Jingyi Zeng
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Chun Zou
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China
| | - Zifei Pei
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA
| | - Cuihua Zou
- Guangxi Medical University Cancer Hospital, Nanning 530022, Guangxi, China.
| | - Yingwei Mao
- Department of Biology, Pennsylvania State University, University Park, PA 16802, USA.
| | - Jiefeng Luo
- Department of Neurology, The Second Affiliated Hospital of Guangxi Medical University, Nanning 530007, Guangxi, China.
| |
Collapse
|
84
|
Haruwaka K, Ying Y, Liang Y, Umpierre AD, Yi MH, Kremen V, Chen T, Xie T, Qi F, Zhao S, Zheng J, Liu YU, Dong H, Worrell GA, Wu LJ. Microglia enhance post-anesthesia neuronal activity by shielding inhibitory synapses. Nat Neurosci 2024; 27:449-461. [PMID: 38177340 PMCID: PMC10960525 DOI: 10.1038/s41593-023-01537-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 11/29/2023] [Indexed: 01/06/2024]
Abstract
Microglia are resident immune cells of the central nervous system and play key roles in brain homeostasis. During anesthesia, microglia increase their dynamic process surveillance and interact more closely with neurons. However, the functional significance of microglial process dynamics and neuronal interaction under anesthesia is largely unknown. Using in vivo two-photon imaging in mice, we show that microglia enhance neuronal activity after the cessation of isoflurane anesthesia. Hyperactive neuron somata are contacted directly by microglial processes, which specifically colocalize with GABAergic boutons. Electron-microscopy-based synaptic reconstruction after two-photon imaging reveals that, during anesthesia, microglial processes enter into the synaptic cleft to shield GABAergic inputs. Microglial ablation or loss of microglial β2-adrenergic receptors prevents post-anesthesia neuronal hyperactivity. Our study demonstrates a previously unappreciated function of microglial process dynamics, which enable microglia to transiently boost post-anesthesia neuronal activity by physically shielding inhibitory inputs.
Collapse
Affiliation(s)
| | - Yanlu Ying
- Department of Anesthesiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China.
| | - Yue Liang
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | | | - Min-Hee Yi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Vaclav Kremen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Tingjun Chen
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Tao Xie
- Translational Oncology Group, Faculty of Science, School of Life Sciences, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Fangfang Qi
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Shunyi Zhao
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Jiaying Zheng
- Department of Neurology, Mayo Clinic, Rochester, MN, USA
| | - Yong U Liu
- Department of Anesthesiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | - Long-Jun Wu
- Department of Neurology, Mayo Clinic, Rochester, MN, USA.
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
85
|
Rifat A, Ossola B, Bürli RW, Dawson LA, Brice NL, Rowland A, Lizio M, Xu X, Page K, Fidzinski P, Onken J, Holtkamp M, Heppner FL, Geiger JRP, Madry C. Differential contribution of THIK-1 K + channels and P2X7 receptors to ATP-mediated neuroinflammation by human microglia. J Neuroinflammation 2024; 21:58. [PMID: 38409076 PMCID: PMC10895799 DOI: 10.1186/s12974-024-03042-6] [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: 11/16/2023] [Accepted: 02/12/2024] [Indexed: 02/28/2024] Open
Abstract
Neuroinflammation is highly influenced by microglia, particularly through activation of the NLRP3 inflammasome and subsequent release of IL-1β. Extracellular ATP is a strong activator of NLRP3 by inducing K+ efflux as a key signaling event, suggesting that K+-permeable ion channels could have high therapeutic potential. In microglia, these include ATP-gated THIK-1 K+ channels and P2X7 receptors, but their interactions and potential therapeutic role in the human brain are unknown. Using a novel specific inhibitor of THIK-1 in combination with patch-clamp electrophysiology in slices of human neocortex, we found that THIK-1 generated the main tonic K+ conductance in microglia that sets the resting membrane potential. Extracellular ATP stimulated K+ efflux in a concentration-dependent manner only via P2X7 and metabotropic potentiation of THIK-1. We further demonstrated that activation of P2X7 was mandatory for ATP-evoked IL-1β release, which was strongly suppressed by blocking THIK-1. Surprisingly, THIK-1 contributed only marginally to the total K+ conductance in the presence of ATP, which was dominated by P2X7. This suggests a previously unknown, K+-independent mechanism of THIK-1 for NLRP3 activation. Nuclear sequencing revealed almost selective expression of THIK-1 in human brain microglia, while P2X7 had a much broader expression. Thus, inhibition of THIK-1 could be an effective and, in contrast to P2X7, microglia-specific therapeutic strategy to contain neuroinflammation.
Collapse
Affiliation(s)
- Ali Rifat
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Bernardino Ossola
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Roland W Bürli
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Lee A Dawson
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Nicola L Brice
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Anna Rowland
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Marina Lizio
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Xiao Xu
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Keith Page
- Cerevance Ltd, 418 Cambridge Science Park, Milton Road, Cambridge, CB4 0PZ, UK
| | - Pawel Fidzinski
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurology, Epilepsy-Center Berlin-Brandenburg, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Neurocure Cluster of Excellence, Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Julia Onken
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neurosurgery, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Martin Holtkamp
- Department of Neurology, Epilepsy-Center Berlin-Brandenburg, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Frank L Heppner
- Neurocure Cluster of Excellence, Neuroscience Clinical Research Center, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- Department of Neuropathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE) Berlin, 10117, Berlin, Germany
| | - Jörg R P Geiger
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany
| | - Christian Madry
- Institute of Neurophysiology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Charitéplatz 1, 10117, Berlin, Germany.
| |
Collapse
|
86
|
Cuní-López C, Stewart R, Oikari LE, Nguyen TH, Roberts TL, Sun Y, Guo CC, Lupton MK, White AR, Quek H. Advanced patient-specific microglia cell models for pre-clinical studies in Alzheimer's disease. J Neuroinflammation 2024; 21:50. [PMID: 38365833 PMCID: PMC10870454 DOI: 10.1186/s12974-024-03037-3] [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: 12/17/2023] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
BACKGROUND Alzheimer's disease (AD) is an incurable neurodegenerative disorder with a rapidly increasing prevalence worldwide. Current approaches targeting hallmark pathological features of AD have had no consistent clinical benefit. Neuroinflammation is a major contributor to neurodegeneration and hence, microglia, the brain's resident immune cells, are an attractive target for potentially more effective therapeutic strategies. However, there is no current in vitro model system that captures AD patient-specific microglial characteristics using physiologically relevant and experimentally flexible culture conditions. METHODS To address this shortcoming, we developed novel 3D Matrigel-based monocyte-derived microglia-like cell (MDMi) mono-cultures and co-cultures with neuro-glial cells (ReNcell VM). We used single-cell RNA sequencing (scRNAseq) analysis to compare the transcriptomic signatures of MDMi between model systems (2D, 3D and 3D co-culture) and against published human microglia datasets. To demonstrate the potential of MDMi for use in personalized pre-clinical strategies, we generated and characterized MDMi models from sixteen AD patients and matched healthy controls, and profiled cytokine responses upon treatment with anti-inflammatory drugs (dasatinib and spiperone). RESULTS MDMi in 3D exhibited a more branched morphology and longer survival in culture compared to 2D. scRNAseq uncovered distinct MDMi subpopulations that exhibit higher functional heterogeneity and best resemble human microglia in 3D co-culture. AD MDMi in 3D co-culture showed altered cell-to-cell interactions, growth factor and cytokine secretion profiles and responses to amyloid-β. Drug testing assays revealed patient- and model-specific cytokine responses. CONCLUSION Our study presents a novel, physiologically relevant and AD patient-specific 3D microglia cell model that opens avenues towards improving personalized drug development strategies in AD.
Collapse
Affiliation(s)
- Carla Cuní-López
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4006, Australia
| | - Romal Stewart
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- UQ Centre for Clinical Research, The University of Queensland, Brisbane City, QLD, 4029, Australia
| | - Lotta E Oikari
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane City, QLD, 4000, Australia
| | - Tam Hong Nguyen
- Scientific Services, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Tara L Roberts
- UQ Centre for Clinical Research, The University of Queensland, Brisbane City, QLD, 4029, Australia
- Ingham Institute for Applied Medical Research and School of Medicine, Western Sydney University, Liverpool, NSW, 2170, Australia
| | - Yifan Sun
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
| | - Christine C Guo
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- ActiGraph LLC, Pensacola, FL, 32502, USA
| | - Michelle K Lupton
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4006, Australia
- School of Biomedical Sciences, Queensland University of Technology, Brisbane City, QLD, 4000, Australia
| | - Anthony R White
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- Faculty of Medicine, The University of Queensland, Herston, QLD, 4006, Australia.
| | - Hazel Quek
- Mental Health and Neuroscience Department, QIMR Berghofer Medical Research Institute, Herston, QLD, 4006, Australia.
- School of Biomedical Sciences, The University of Queensland, Lucia, QLD, 4072, Australia.
- School of Biomedical Sciences, Queensland University of Technology, Brisbane City, QLD, 4000, Australia.
| |
Collapse
|
87
|
Zhao S, Wang L, Liang Y, Zheng J, Umpierre AD, Wu LJ. Chemogenetic activation of microglial Gi signaling decreases microglial surveillance and impairs neuronal synchronization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.12.579861. [PMID: 38405754 PMCID: PMC10888941 DOI: 10.1101/2024.02.12.579861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Microglia actively survey the brain and dynamically interact with neurons to maintain brain homeostasis. Microglial Gi-protein coupled receptors (Gi-GPCRs) play a critical role in microglia-neuron communications. However, the impact of temporally activating microglial Gi signaling on microglial dynamics and neuronal activity in the homeostatic brain remains largely unknown. In this study, we employed Gi-based Designer Receptors Exclusively Activated by Designer Drugs (Gi-DREADD) to selectively and temporally modulate microglial Gi signaling pathway. By integrating this chemogenetic approach with in vivo two-photon imaging, we observed that exogenous activation of microglial Gi signaling transiently inhibited microglial process dynamics, reduced neuronal activity, and impaired neuronal synchronization. These altered neuronal functions were associated with a decrease in interactions between microglia and neuron somata. Altogether, this study demonstrates that acute, exogenous activation of microglial Gi signaling can regulate neuronal circuit function, offering a potential pharmacological target for neuromodulation through microglia.
Collapse
|
88
|
Denes A, Hansen CE, Oezorhan U, Figuerola S, de Vries HE, Sorokin L, Planas AM, Engelhardt B, Schwaninger M. Endothelial cells and macrophages as allies in the healthy and diseased brain. Acta Neuropathol 2024; 147:38. [PMID: 38347307 PMCID: PMC10861611 DOI: 10.1007/s00401-024-02695-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 01/19/2024] [Accepted: 01/20/2024] [Indexed: 02/15/2024]
Abstract
Diseases of the central nervous system (CNS) are often associated with vascular disturbances or inflammation and frequently both. Consequently, endothelial cells and macrophages are key cellular players that mediate pathology in many CNS diseases. Macrophages in the brain consist of the CNS-associated macrophages (CAMs) [also referred to as border-associated macrophages (BAMs)] and microglia, both of which are close neighbours or even form direct contacts with endothelial cells in microvessels. Recent progress has revealed that different macrophage populations in the CNS and a subset of brain endothelial cells are derived from the same erythromyeloid progenitor cells. Macrophages and endothelial cells share several common features in their life cycle-from invasion into the CNS early during embryonic development and proliferation in the CNS, to their demise. In adults, microglia and CAMs have been implicated in regulating the patency and diameter of vessels, blood flow, the tightness of the blood-brain barrier, the removal of vascular calcification, and the life-time of brain endothelial cells. Conversely, CNS endothelial cells may affect the polarization and activation state of myeloid populations. The molecular mechanisms governing the pas de deux of brain macrophages and endothelial cells are beginning to be deciphered and will be reviewed here.
Collapse
Affiliation(s)
- Adam Denes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Cathrin E Hansen
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Uemit Oezorhan
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany
| | - Sara Figuerola
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomedicas de Barcelona (IIBB), Consejo Superior de Investigaciones Cientificas (CSIC), 08036, Barcelona, Spain
- Cerebrovascular Research Group, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam UMC Location Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Amsterdam Neuroscience, Amsterdam UMC, Amsterdam, The Netherlands
- MS Center Amsterdam, Amsterdam UMC Location VU Medical Center, Amsterdam, The Netherlands
| | - Lydia Sorokin
- Institute of Physiological Chemistry and Pathobiochemistry, University of Muenster, Munster, Germany
- Cells-in-Motion Interfaculty Centre (CIMIC), University of Münster, Münster, Germany
| | - Anna M Planas
- Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomedicas de Barcelona (IIBB), Consejo Superior de Investigaciones Cientificas (CSIC), 08036, Barcelona, Spain
- Cerebrovascular Research Group, Institut d'Investigacions Biomediques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
- Faculty of Medicine, University of Barcelona, Barcelona, Spain
| | | | - Markus Schwaninger
- Institute of Experimental and Clinical Pharmacology and Toxicology, Center of Brain, Behavior and Metabolism, University of Lübeck, Lübeck, Germany.
- German Research Centre for Cardiovascular Research (DZHK), Partner Site Hamburg, Lübeck, Kiel, Germany.
| |
Collapse
|
89
|
Di Martino E, Rayasam A, Vexler ZS. Brain Maturation as a Fundamental Factor in Immune-Neurovascular Interactions in Stroke. Transl Stroke Res 2024; 15:69-86. [PMID: 36705821 PMCID: PMC10796425 DOI: 10.1007/s12975-022-01111-7] [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: 07/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 01/28/2023]
Abstract
Injuries in the developing brain cause significant long-term neurological deficits. Emerging clinical and preclinical data have demonstrated that the pathophysiology of neonatal and childhood stroke share similar mechanisms that regulate brain damage, but also have distinct molecular signatures and cellular pathways. The focus of this review is on two different diseases-neonatal and childhood stroke-with emphasis on similarities and distinctions identified thus far in rodent models of these diseases. This includes the susceptibility of distinct cell types to brain injury with particular emphasis on the role of resident and peripheral immune populations in modulating stroke outcome. Furthermore, we discuss some of the most recent and relevant findings in relation to the immune-neurovascular crosstalk and how the influence of inflammatory mediators is dependent on specific brain maturation stages. Finally, we comment on the current state of treatments geared toward inducing neuroprotection and promoting brain repair after injury and highlight that future prophylactic and therapeutic strategies for stroke should be age-specific and consider gender differences in order to achieve optimal translational success.
Collapse
Affiliation(s)
- Elena Di Martino
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Aditya Rayasam
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA
| | - Zinaida S Vexler
- Department of Neurology, University California San Francisco, 675 Nelson Rising Lane, San Francisco, CA, 94158-0663, USA.
| |
Collapse
|
90
|
Sahara N, Higuchi M. Diagnostic and therapeutic targeting of pathological tau proteins in neurodegenerative disorders. FEBS Open Bio 2024; 14:165-180. [PMID: 37746832 PMCID: PMC10839408 DOI: 10.1002/2211-5463.13711] [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: 07/24/2023] [Revised: 09/06/2023] [Accepted: 09/22/2023] [Indexed: 09/26/2023] Open
Abstract
Tauopathies, characterized by fibrillar tau accumulation in neurons and glial cells, constitute a major neuropathological category of neurodegenerative diseases. Neurofibrillary tau lesions are strongly associated with cognitive deficits in these diseases, but the causal mechanisms underlying tau-induced neuronal dysfunction remain unresolved. Recent advances in cryo-electron microscopy examination have revealed various core structures of tau filaments from different tauopathy patients, which can be used to classify tauopathies. In vivo visualization of tau pathology is now available using several tau positron emission tomography tracers. Among these radioprobes, PM-PBB3 allows high-contrast imaging of tau deposits in the brains of patients with diverse disorders and tauopathy mouse models. Selective degradation of pathological tau species by the ubiquitin-proteasome system or autophagy machinery is a potential therapeutic strategy. Alternatively, the non-cell-autonomous clearance of pathological tau species through neuron-glia networks could be reinforced as a disease-modifying treatment. In addition, the development of neuroinflammatory biomarkers is required for understanding the contribution of immunocompetent cells in the brain to preventing neurodegeneration. This review provides an overview of the current research and development of diagnostic and therapeutic agents targeting divergent tau pathologies.
Collapse
Affiliation(s)
- Naruhiko Sahara
- Department of Functional Brain Imaging, Institute for Quantum Medical SciencesNational Institutes for Quantum Science and TechnologyChibaJapan
| | - Makoto Higuchi
- Department of Functional Brain Imaging, Institute for Quantum Medical SciencesNational Institutes for Quantum Science and TechnologyChibaJapan
| |
Collapse
|
91
|
Leek AN, Quinn JA, Krapf D, Tamkun MM. GLT-1a glutamate transporter nanocluster localization is associated with astrocytic actin and neuronal Kv2 clusters at sites of neuron-astrocyte contact. Front Cell Dev Biol 2024; 12:1334861. [PMID: 38362041 PMCID: PMC10867268 DOI: 10.3389/fcell.2024.1334861] [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: 11/07/2023] [Accepted: 01/16/2024] [Indexed: 02/17/2024] Open
Abstract
Introduction: Astrocytic GLT-1 glutamate transporters ensure the fidelity of glutamic neurotransmission by spatially and temporally limiting glutamate signals. The ability to limit neuronal hyperactivity relies on the localization and diffusion of GLT-1 on the astrocytic surface, however, little is known about the underlying mechanisms. We show that two isoforms of GLT-1, GLT-1a and GLT-1b, form nanoclusters on the surface of transfected astrocytes and HEK-293 cells. Methods: We used both fixed and live cell super-resolution imaging of fluorescent protein and epitope tagged proteins in co-cultures of rat astrocytes and neurons. Immunofluorescence techniques were also used. GLT1 diffusion was assessed via single particle tracking and fluorescence recovery after photobleach (FRAP). Results: We found GLT-1a, but not GLT-1b, nanoclusters concentrated adjacent to actin filaments which was maintained after addition of glutamate. GLT-1a nanocluster concentration near actin filaments was prevented by expression of a cytosolic GLT-1a C-terminus, suggesting the C-terminus is involved in the localization adjacent to cortical actin. Using super-resolution imaging, we show that astrocytic GLT-1a and actin co-localize in net-like structures around neuronal Kv2.1 clusters at points of neuron/astrocyte contact. Conclusion: Overall, these data describe a novel relationship between GLT-1a and cortical actin filaments, which localizes GLT-1a near neuronal structures responsive to ischemic insult.
Collapse
Affiliation(s)
- Ashley N. Leek
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States
| | - Josiah A. Quinn
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
| | - Diego Krapf
- Department of Electrical and Computer Engineering, Colorado State University, Fort Collins, CO, United States
| | - Michael M. Tamkun
- Department of Biomedical Sciences, Colorado State University, Fort Collins, CO, United States
- Molecular, Cellular and Integrative Neuroscience Program, Colorado State University, Fort Collins, CO, United States
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO, United States
| |
Collapse
|
92
|
Ma C, Li B, Silverman D, Ding X, Li A, Xiao C, Huang G, Worden K, Muroy S, Chen W, Xu Z, Tso CF, Huang Y, Zhang Y, Luo Q, Saijo K, Dan Y. Microglia regulate sleep through calcium-dependent modulation of norepinephrine transmission. Nat Neurosci 2024; 27:249-258. [PMID: 38238430 PMCID: PMC10849959 DOI: 10.1038/s41593-023-01548-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 12/08/2023] [Indexed: 02/09/2024]
Abstract
Sleep interacts reciprocally with immune system activity, but its specific relationship with microglia-the resident immune cells in the brain-remains poorly understood. Here, we show in mice that microglia can regulate sleep through a mechanism involving Gi-coupled GPCRs, intracellular Ca2+ signaling and suppression of norepinephrine transmission. Chemogenetic activation of microglia Gi signaling strongly promoted sleep, whereas pharmacological blockade of Gi-coupled P2Y12 receptors decreased sleep. Two-photon imaging in the cortex showed that P2Y12-Gi activation elevated microglia intracellular Ca2+, and blockade of this Ca2+ elevation largely abolished the Gi-induced sleep increase. Microglia Ca2+ level also increased at natural wake-to-sleep transitions, caused partly by reduced norepinephrine levels. Furthermore, imaging of norepinephrine with its biosensor in the cortex showed that microglia P2Y12-Gi activation significantly reduced norepinephrine levels, partly by increasing the adenosine concentration. These findings indicate that microglia can regulate sleep through reciprocal interactions with norepinephrine transmission.
Collapse
Affiliation(s)
- Chenyan Ma
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Bing Li
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Daniel Silverman
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Xinlu Ding
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Anan Li
- Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainmatics, JITRI, Suzhou, China
| | - Chi Xiao
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Ganghua Huang
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Kurtresha Worden
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Sandra Muroy
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Wei Chen
- Department of Physics, University of California, Berkeley, Berkeley, CA, USA
| | - Zhengchao Xu
- Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
| | - Chak Foon Tso
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
- , Sunnyvale, CA, USA
| | - Yixuan Huang
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Yufan Zhang
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Qingming Luo
- Biomedical Photonics, Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainmatics, JITRI, Suzhou, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Kaoru Saijo
- Division of Immunology and Pathogenesis, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| | - Yang Dan
- Division of Neurobiology, Department of Molecular and Cell Biology, Helen Wills Neuroscience Institute, Howard Hughes Medical Institute, University of California, Berkeley, Berkeley, CA, USA.
| |
Collapse
|
93
|
Shen QQ, Jv XH, Ma XZ, Li C, Liu L, Jia WT, Qu L, Chen LL, Xie JX. Cell senescence induced by toxic interaction between α-synuclein and iron precedes nigral dopaminergic neuron loss in a mouse model of Parkinson's disease. Acta Pharmacol Sin 2024; 45:268-281. [PMID: 37674042 PMCID: PMC10789811 DOI: 10.1038/s41401-023-01153-z] [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: 04/07/2023] [Accepted: 08/10/2023] [Indexed: 09/08/2023] Open
Abstract
Cell senescence has been implicated in the pathology of Parkinson's disease (PD). Both abnormal α-synuclein aggregation and iron deposition are suggested to be the triggers, facilitators, and aggravators during the development of PD. In this study, we investigated the involvement of α-synuclein and iron in the process of cell senescence in a mouse model of PD. In order to overexpress α-syn-A53T in the substantia nigra pars compacta (SNpc), human α-syn-A53T was microinjected into both sides of the SNpc in mice. We found that overexpression of α-syn-A53T for one week induced significant pro-inflammatory senescence-associated secretory phenotype (SASP), increased cell senescence-related proteins (β-gal, p16, p21, H2A.X and γ-H2A.X), mitochondrial dysfunction accompanied by dysregulation of iron-related proteins (L-ferritin, H-ferritin, DMT1, IRP1 and IRP2) in the SNpc. In contrast, significant loss of nigral dopaminergic neurons and motor dysfunction were only observed after overexpression of α-syn-A53T for 4 weeks. In PC12 cells stably overexpressing α-syn-A53T, iron overload (ferric ammonium citrate, FAC, 100 μM) not only increased the level of reactive oxygen species (ROS), p16 and p21, but also exacerbated the processes of oxidative stress and cell senescence signalling induced by α-syn-A53T overexpression. Interestingly, reducing the iron level with deferoxamine (DFO) or knockdown of transferrin receptor 1 (TfR1) significantly improved both the phenotypes and dysregulated proteins of cell senescence induced by α-syn-A53T overexpression. All these evidence highlights the toxic interaction between iron and α-synuclein inducing cell senescence, which precedes nigral dopaminergic neuronal loss in PD. Further investigation on cell senescence may yield new therapeutic agents for the prevention or treatment of PD.
Collapse
Affiliation(s)
- Qing-Qing Shen
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Xian-Hui Jv
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Xi-Zhen Ma
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Chong Li
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Lin Liu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Wen-Ting Jia
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Le Qu
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China
| | - Lei-Lei Chen
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China.
| | - Jun-Xia Xie
- Institute of Brain Science and Disease, Shandong Provincial Collaborative Innovation Center for Neurodegenerative Disorders, Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders, Qingdao University, Qingdao, 266021, China.
| |
Collapse
|
94
|
Granzotto A, McQuade A, Chadarevian JP, Davtyan H, Sensi SL, Parker I, Blurton-Jones M, Smith I. ER and SOCE Ca 2+ signals are not required for directed cell migration in human microglia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.18.576126. [PMID: 38293075 PMCID: PMC10827168 DOI: 10.1101/2024.01.18.576126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2024]
Abstract
The central nervous system (CNS) is constantly surveilled by microglia, highly motile and dynamic cells deputed to act as the first line of immune defense in the brain and spinal cord. Alterations in the homeostasis of the CNS are detected by microglia that respond by migrating toward the affected area. Understanding the mechanisms controlling directed cell migration of microglia is crucial to dissect their responses to neuroinflammation and injury. We used a combination of pharmacological and genetic approaches to explore the involvement of calcium (Ca2+) signaling in the directed migration of induced pluripotent stem cell (iPSC)-derived microglia challenged with a purinergic stimulus. This approach mimics cues originating from injury of the CNS. Unexpectedly, simultaneous imaging of microglia migration and intracellular Ca2+ changes revealed that this phenomenon does not require Ca2+ signals generated from the endoplasmic reticulum (ER) and store-operated Ca2+ entry (SOCE) pathways. Instead, we find evidence that human microglial chemotaxis to purinergic signals is mediated by cyclic AMP in a Ca2+-independent manner. These results challenge prevailing notions, with important implications in neurological conditions characterized by perturbation in Ca2+ homeostasis.
Collapse
Affiliation(s)
- Alberto Granzotto
- UCI Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, United States
- Center for Advanced Sciences and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, University G d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Amanda McQuade
- UCI Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, United States
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, United States
| | - Jean Paul Chadarevian
- UCI Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, United States
| | - Hayk Davtyan
- UCI Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, United States
| | - Stefano L. Sensi
- Center for Advanced Sciences and Technology (CAST), University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, University G d’Annunzio of Chieti-Pescara, Chieti, Italy
- Institute for Advanced Biomedical Technologies (ITAB), “G. d’Annunzio” University, Chieti-Pescara, Italy
| | - Ian Parker
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, United States
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, United States
| | - Mathew Blurton-Jones
- UCI Institute for Memory Impairments and Neurological Disorders, University of California, Irvine, United States
- Sue and Bill Gross Stem Cell Research Center, University of California, Irvine, Irvine, United States
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, United States
- Institute for Immunology, University of California, Irvine, Irvine, United States
| | - Ian Smith
- Department of Neurobiology and Behavior, University of California, Irvine, Irvine, United States
| |
Collapse
|
95
|
Shigetomi E, Sakai K, Koizumi S. Extracellular ATP/adenosine dynamics in the brain and its role in health and disease. Front Cell Dev Biol 2024; 11:1343653. [PMID: 38304611 PMCID: PMC10830686 DOI: 10.3389/fcell.2023.1343653] [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: 11/24/2023] [Accepted: 12/31/2023] [Indexed: 02/03/2024] Open
Abstract
Extracellular ATP and adenosine are neuromodulators that regulate numerous neuronal functions in the brain. Neuronal activity and brain insults such as ischemic and traumatic injury upregulate these neuromodulators, which exert their effects by activating purinergic receptors. In addition, extracellular ATP/adenosine signaling plays a pivotal role in the pathogenesis of neurological diseases. Virtually every cell type in the brain contributes to the elevation of ATP/adenosine, and various mechanisms underlying this increase have been proposed. Extracellular adenosine is thought to be mainly produced via the degradation of extracellular ATP. However, adenosine is also released from neurons and glia in the brain. Therefore, the regulation of extracellular ATP/adenosine in physiological and pathophysiological conditions is likely far more complex than previously thought. To elucidate the complex mechanisms that regulate extracellular ATP/adenosine levels, accurate methods of assessing their spatiotemporal dynamics are needed. Several novel techniques for acquiring spatiotemporal information on extracellular ATP/adenosine, including fluorescent sensors, have been developed and have started to reveal the mechanisms underlying the release, uptake and degradation of ATP/adenosine. Here, we review methods for analyzing extracellular ATP/adenosine dynamics as well as the current state of knowledge on the spatiotemporal dynamics of ATP/adenosine in the brain. We focus on the mechanisms used by neurons and glia to cooperatively produce the activity-dependent increase in ATP/adenosine and its physiological and pathophysiological significance in the brain.
Collapse
Affiliation(s)
- Eiji Shigetomi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
- Yamanashi GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Kent Sakai
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
- Yamanashi GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
- Yamanashi GLIA Center, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Japan
| |
Collapse
|
96
|
Jiang S, Ma Y, Shi Y, Zou Y, Yang Z, Zhi W, Zhao Z, Shen W, Chen L, Wu Y, Wang L, Hu X, Wu H. Acute exposure of microwave impairs attention process by activating microglial inflammation. Cell Biosci 2024; 14:2. [PMID: 38178181 PMCID: PMC10768366 DOI: 10.1186/s13578-023-01162-9] [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: 07/12/2023] [Accepted: 11/02/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND Attention provides the foundation for cognitions, which was shown to be affected by microwave (MW) radiation. With the ubiquitous of microwaves, public concerns regarding the impact of MW radiation on attention has hence been increased. Our study aims to investigate the potential effect and mechanism of acute microwave exposure on attention. RESULTS We identified obvious impairment of attention in mice by the five-choice serial reaction time (5-CSRT) task. Proteomic analysis of the cerebrospinal fluid (CSF) revealed neuroinflammation and microglial activation potentially due to acute MW exposure. Moreover, biochemical analysis further confirmed microglial activation in the prefrontal cortex (PFC) of mice subjected to acute MW exposure. Finally, minocycline, a commercially available anti-inflammatory compound, attenuated neuroinflammation, inhibited the upregulation of N-methyl-D-aspartic acid receptor (NMDAR) including NR2A and NR2B, and also accelerated the attentional recovery after MW exposure. CONCLUSIONS We believe that microglial activation and NMDAR upregulation likely contribute to inattention induced by acute MW exposure, and we found that minocycline may be effective in preventing such process.
Collapse
Affiliation(s)
- Shaofei Jiang
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Yingping Ma
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui Province, China
| | - Yuan Shi
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yong Zou
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhenqi Yang
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Weijia Zhi
- Beijing Institute of Radiation Medicine, Beijing, China
| | - Zhe Zhao
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Wei Shen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Liping Chen
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Yan Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China
| | - Lifeng Wang
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Xiangjun Hu
- Beijing Institute of Radiation Medicine, Beijing, China.
| | - Haitao Wu
- Department of Neurobiology, Beijing Institute of Basic Medical Sciences, Beijing, China.
- School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui Province, China.
- Key Laboratory of Neuroregeneration, Co-Innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu Province, China.
- Chinese Institute for Brain Research, Beijing, China.
| |
Collapse
|
97
|
Cserép C, Pósfai B, Szabadits E, Dénes Á. Contactomics of Microglia and Intercellular Communication. ADVANCES IN NEUROBIOLOGY 2024; 37:135-149. [PMID: 39207690 DOI: 10.1007/978-3-031-55529-9_8] [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
Microglia represent the main immunocompetent cell type in the parenchyma of the brain and the spinal cord, with roles extending way beyond their immune functions. While emerging data show the pivotal role of microglia in brain development, brain health and brain diseases, the exact mechanisms through which microglia contribute to complex neuroimmune interactions are still largely unclear. Understanding the communication between microglia and other cells represents an important cornerstone of these interactions, which may provide novel opportunities for therapeutic interventions in neurological or psychiatric disorders. As such, in line with studying the effects of the numerous soluble mediators that influence neuroimmune processes, attention on physical interactions between microglia and other cells in the CNS has increased substantially in recent years. In this chapter, we briefly summarize the latest literature on "microglial contactomics" and its functional implications in health and disease.
Collapse
Affiliation(s)
- Csaba Cserép
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Balázs Pósfai
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Eszter Szabadits
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary.
| |
Collapse
|
98
|
Augusto-Oliveira M, Tremblay MÈ, Verkhratsky A. Receptors on Microglia. ADVANCES IN NEUROBIOLOGY 2024; 37:83-121. [PMID: 39207688 DOI: 10.1007/978-3-031-55529-9_6] [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
Microglial cells are the most receptive cells in the central nervous system (CNS), expressing several classes of receptors reflecting their immune heritage and newly acquired neural specialisation. Microglia possess, depending on the particular context, receptors to neurotransmitters and neuromodulators as well as immunocompetent receptors. This rich complement allows microglial cells to monitor the functional status of the nervous system, contribute actively to the regulation of neural activity and plasticity and homeostasis, and guard against pathogens as well as other challenges to the CNS's integrity and function.
Collapse
Affiliation(s)
- Marcus Augusto-Oliveira
- Laboratório de Farmacologia Molecular, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
- Programa de Pós-Graduação em Farmacologia e Bioquímica, Instituto de Ciências Biológicas, Universidade Federal do Pará, Belém, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, Medical Sciences Building, University of Victoria, Victoria, BC, Canada
- Axe neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Pavillon Ferdinand-Vandry, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Life Sciences Center, Vancouver, BC, Canada
| | - Alexei Verkhratsky
- Faculty of Life Sciences, The University of Manchester, Manchester, UK.
- Department of Neurosciences, University of the Basque Country, Leioa, Spain.
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain.
| |
Collapse
|
99
|
Kim JS, Jung S. Visualization, Fate Mapping, Ablation, and Mutagenesis of Microglia in the Mouse Brain. ADVANCES IN NEUROBIOLOGY 2024; 37:53-63. [PMID: 39207686 DOI: 10.1007/978-3-031-55529-9_4] [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
Since the classical studies of Pío del Río-Hortega, microglia research has come a long way. In particular, recent advances in bulk and single-cell (sc) transcriptomics have yielded many fascinating new insights into these intriguing immune cells at the interface with the central nervous system (CNS), both in small animal models and human samples. In parallel, tools developed by advanced mouse genetics have revealed the unique ontogeny of microglia and their striking dynamic interactions with other cells in the brain parenchyma. In this chapter, we will discuss various applications of the Cre/loxP-based approach that have enabled the study of microglia in their physiological context of the mouse brain. We will highlight selected key findings that have shaped our current understanding of these cells and discuss the technical intricacies of the Cre/loxP approach and some remaining challenges.
Collapse
Affiliation(s)
- Jung-Seok Kim
- Department of Immunology and Regenerative Biology (IRB), Weizmann Institute of Science, Rehovot, Israel.
| | - Steffen Jung
- Department of Immunology and Regenerative Biology (IRB), Weizmann Institute of Science, Rehovot, Israel
| |
Collapse
|
100
|
Newton K, De Biase L. Substance Use and Addiction. ADVANCES IN NEUROBIOLOGY 2024; 37:343-355. [PMID: 39207701 DOI: 10.1007/978-3-031-55529-9_19] [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
Efforts to reveal the molecular, cellular, and circuit mechanisms of addiction have largely focused on neurons. Yet accumulating data regarding the ability of glial cells to impact synaptic function, circuit activity, and behavior demands that we explore how these nonneuronal cells contribute to substance use disorders and addiction. Important work has shown that glial cells, including microglia, exhibit changes in phenotype following exposure to drugs of abuse and that modification of glial responses can impact behaviors related to drug seeking and drug taking. While these are critical first steps to understanding how microglia can impact addiction, there are still substantial gaps in knowledge that need to be addressed. This chapter reviews some of the key studies that have shown how microglia are affected by and can contribute to addiction. It also discusses areas where more knowledge is urgently needed to reveal new therapeutic and preventative approaches.
Collapse
Affiliation(s)
- Keionna Newton
- Neuroscience Interdepartmental Graduate Program, University of California, Los Angeles, CA, USA
| | - Lindsay De Biase
- Department of Physiology, University of California, Los Angeles, CA, USA.
| |
Collapse
|