1
|
Zha X, Zheng G, Skutella T, Kiening K, Unterberg A, Younsi A. Microglia: a promising therapeutic target in spinal cord injury. Neural Regen Res 2025; 20:454-463. [PMID: 38819048 PMCID: PMC11317945 DOI: 10.4103/nrr.nrr-d-23-02044] [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: 12/18/2023] [Revised: 02/23/2024] [Accepted: 03/22/2024] [Indexed: 06/01/2024] Open
Abstract
Microglia are present throughout the central nervous system and are vital in neural repair, nutrition, phagocytosis, immunological regulation, and maintaining neuronal function. In a healthy spinal cord, microglia are accountable for immune surveillance, however, when a spinal cord injury occurs, the microenvironment drastically changes, leading to glial scars and failed axonal regeneration. In this context, microglia vary their gene and protein expression during activation, and proliferation in reaction to the injury, influencing injury responses both favorably and unfavorably. A dynamic and multifaceted injury response is mediated by microglia, which interact directly with neurons, astrocytes, oligodendrocytes, and neural stem/progenitor cells. Despite a clear understanding of their essential nature and origin, the mechanisms of action and new functions of microglia in spinal cord injury require extensive research. This review summarizes current studies on microglial genesis, physiological function, and pathological state, highlights their crucial roles in spinal cord injury, and proposes microglia as a therapeutic target.
Collapse
Affiliation(s)
- Xiaowei Zha
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Guoli Zheng
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Thomas Skutella
- Department of Neuroanatomy, Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Karl Kiening
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Andreas Unterberg
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Younsi
- Department of Neurosurgery, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
2
|
Bizen N, Takebayashi H. Diverse functions of DEAD-box proteins in oligodendrocyte development, differentiation, and homeostasis. J Neurochem 2025; 169:e16238. [PMID: 39374171 DOI: 10.1111/jnc.16238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/18/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024]
Abstract
Oligodendrocytes, a type of glial cell in the central nervous system, have a critical role in the formation of myelin around axons, facilitating saltatory conduction, and maintaining the integrity of nerve axons. The dysregulation of oligodendrocyte differentiation and homeostasis have been implicated in a wide range of neurological diseases, including dysmyelinating disorders (e.g., Pelizaeus-Merzbacher disease), demyelinating diseases (e.g., multiple sclerosis), Alzheimer's disease, and psychiatric disorders. Therefore, unraveling the mechanisms of oligodendrocyte development, differentiation, and homeostasis is essential for understanding the pathogenesis of these diseases and the development of therapeutic interventions. Numerous studies have identified and analyzed the functions of transcription factors, RNA metabolic factors, translation control factors, and intracellular and extracellular signals involved in the series of processes from oligodendrocyte fate determination to terminal differentiation. DEAD-box proteins, multifunctional RNA helicases that regulate various intracellular processes, including transcription, RNA processing, and translation, are increasingly recognized for their diverse roles in various aspects of oligodendrocyte development, differentiation, and maintenance of homeostasis. This review introduces the latest insights into the regulatory networks of oligodendrocyte biology mediated by DEAD-box proteins.
Collapse
Affiliation(s)
- Norihisa Bizen
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
- Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
Zhang H, Yang S, Lu YL, Zhou LQ, Dong MH, Chu YH, Pang XW, Chen L, Xu LL, Zhang LY, Zhu LF, Xu T, Wang W, Shang K, Tian DS, Qin C. Microglial Nrf2-mediated lipid and iron metabolism reprogramming promotes remyelination during white matter ischemia. Redox Biol 2024; 79:103473. [PMID: 39718294 DOI: 10.1016/j.redox.2024.103473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 12/06/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Oxidative stress and microglial activation are critical pathomechanisms in ischemic white matter injury. Microglia, as resident immune cells in the brain, are the main cells undergoing oxidative stress response. However, the role and molecular mechanism of oxidative stress in microglia have not been clearly elucidated during white matter ischemia. METHODS Extensive histological analysis of the corpus callosum was performed in BCAS mice at different time points to assess white matter injury, oxidative stress and microglial activation. Flow cytometric sorting and transcriptomic sequencing were combined to explore the underlying mechanisms regulating microglial oxidative stress and functional phenotypes. The expression of critical molecule in microglia was regulated using Cx3cr1CreER mice and clinical-stage drugs to assess its effect on white matter injury and cognitive function. RESULTS Our study identified nuclear factor erythroid-2 related factor 2 (Nrf2) as a key transcription factor regulating oxidative stress and functional phenotype in microglia. Interestingly, we found that the sustained decrease in transiently upregulated expression of Nrf2 following chronic cerebral hypoperfusion resulted in abnormal microglial activation and white matter injury. In addition, high loads of myelin debris promoted lipid peroxidation and ferroptosis in microglia with diminished antioxidant function. Microglia with pharmacologically or genetically stimulated Nrf2 expression exhibited enhanced resistance to ferroptosis and pro-regenerative properties to myelination due to lipid and iron metabolism reprogramming. CONCLUSION Weakened Nrf2-mediated antioxidant responses in microglia induced metabolic disturbances and ferroptosis during chronic cerebral hypoperfusion. Targeted enhancement of Nrf2 expression in microglia may be a potential therapeutic strategy for ischemic white matter injury.
Collapse
Affiliation(s)
- Hang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Sheng Yang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yi-Lin Lu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Luo-Qi Zhou
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ming-Hao Dong
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Yun-Hui Chu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Xiao-Wei Pang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Lian Chen
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Lu-Lu Xu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Lu-Yang Zhang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Li-Fang Zhu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ting Xu
- Guangdong Province Key Laboratory of Brain Function and Disease, Department of Physiology and Pain Research Center, Zhongshan Medical School, Sun Yat-sen University, 74 Zhongshan Road 2, Guangzhou, 510080, PR China
| | - Wei Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China
| | - Ke Shang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Dai-Shi Tian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| | - Chuan Qin
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Hubei Key Laboratory of Neural Injury and Functional Reconstruction, Huazhong University of Science and Technology, Wuhan, 430030, PR China; Key Laboratory of Vascular Aging, Ministry of Education, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, PR China.
| |
Collapse
|
4
|
Zhu H, Hu E, Guo X, Yuan Z, Jiang H, Zhang W, Tang T, Wang Y, Li T. Promoting remyelination in central nervous system diseases: Potentials and prospects of natural products and herbal medicine. Pharmacol Res 2024; 210:107533. [PMID: 39617281 DOI: 10.1016/j.phrs.2024.107533] [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: 10/09/2024] [Revised: 11/22/2024] [Accepted: 11/28/2024] [Indexed: 12/20/2024]
Abstract
Myelin damage is frequently associated with central nervous system (CNS) diseases and is a critical factor influencing neurological function and disease prognosis. Nevertheless, the majority of current treatments for the CNS concentrate on gray matter injury and repair strategies, while clinical interventions specifically targeting myelin repair remain unavailable. In recent years, natural products and herbal medicine have achieved considerable progress in the domain of myelin repair, given their remarkable curative effect and low toxic side effects, demonstrating significant therapeutic potential. In this review, we present a rather comprehensive account of the mechanisms underlying myelin formation, injury, and repair, with a particular emphasis on the interactions between oligodendrocytes and other glial cells. Furthermore, we summarize the natural products and herbal medicine currently employed in remyelination along with their mechanisms of action, highlighting the potential and challenges of certain natural compounds to enhance myelin repair. This review aims to facilitate the expedited development of innovative therapeutics derived from natural products and herbal medicine and furnish novel insights into myelin repair in the CNS.
Collapse
Affiliation(s)
- Haonan Zhu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - En Hu
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Xin Guo
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Zhiqiang Yuan
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Haoying Jiang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Wei Zhang
- The College of Integrated Traditional Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
| | - Tao Tang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Yang Wang
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China
| | - Teng Li
- Institute of Integrative Chinese Medicine, Department of Integrated Chinese Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Hunan Key Laboratory of TCM Gan, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China; Department of Neurology of Integrated Chinese Medicine, Xiangya Jiangxi Hospital, Central South University, Nanchang 330006, PR China.
| |
Collapse
|
5
|
Haque A, Zaman V, Drasites KP, Matzelle D, Sawant S, Vertegel A, Varma A, Banik NL. Induction of Neural Differentiation and Protection by a Novel Slow-Release Nanoparticle Estrogen Construct in a Rat Model of Spinal Cord Injury. Neurochem Res 2024; 50:41. [PMID: 39613948 DOI: 10.1007/s11064-024-04289-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 12/01/2024]
Abstract
Spinal cord injury (SCI) is a complex debilitating condition leading to permanent life-long neurological deficits. Estrogen (E2) treatment is known to be neuroprotectant in SCI. This hormone is highly pleiotropic and has been shown to decrease apoptosis, modulate calcium signaling, regulate growth factor expression, act as an anti-inflammatory, and drive angiogenesis. These beneficial effects were found in our earlier study at the low dose of 10 µg/kg E2 in rats. However, the dose remains non-physiologic, which poses a safety hurdle for clinical use. Thus, we recently devised/constructed a fast release nanoparticle (NP) estrogen embedded (FNP-E2) construct and tested a focal delivery system in a contused SCI rat model which showed protection in the short run. In the current study, we have developed a novel slow-release NP estrogen (SNP-E2) delivery system that shows sustained release of E2 in the injured spinal cord and no systemic exposure in the host. The study of E2 release and kinetics of this SNP-E2 construct in vitro and in vivo supported this claim. Delivery of E2 to the injured spinal cord via this approach reduced inflammation and gliosis, and induced microglial differentiation of M1 to M2 in rats after SCI. Analysis of spinal cord samples showed improved myelination and survival signals (AKT) as demonstrated by western blot analysis. SNP-E2 treatment also induced astrocytic differentiation into neuron-like (MAP2/NeuN) cells, supported the survival of oligodendrocyte precursor cells (OPC), and improved bladder and locomotor function in rats following SCI. These data suggest that this novel delivery strategy of SNP-E2 to the injured spinal cord may provide a safe and effective therapeutic approach to treat individuals suffering from SCI.
Collapse
Affiliation(s)
- Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
| | - Kelsey P Drasites
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA
| | - Sushant Sawant
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Alexey Vertegel
- Department of Bioengineering, Clemson University, Clemson, SC, USA
| | - Abhay Varma
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA
| | - Naren L Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC, 29425, USA.
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas Street, Charleston, SC, 29425, USA.
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St, Charleston, SC, 29401, USA.
| |
Collapse
|
6
|
Xu Y, Zhang E, Wei L, Dai Z, Chen S, Zhou S, Huang Y. NINJ1: A new player in multiple sclerosis pathogenesis and potential therapeutic target. Int Immunopharmacol 2024; 141:113021. [PMID: 39197295 DOI: 10.1016/j.intimp.2024.113021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 08/07/2024] [Accepted: 08/23/2024] [Indexed: 09/01/2024]
Abstract
Multiple sclerosis (MS) is a chronic inflammatory disease of the central nervous system (CNS) characterized by demyelination. Current treatment options for MS focus on immunosuppression, but their efficacy can be limited. Recent studies suggest a potential role for nerve injury-induced protein 1 (NINJ1) in MS pathogenesis. NINJ1, a protein involved in cell death and inflammation, may contribute to the infiltration and activation of inflammatory cells in the CNS, potentially through enhanced blood-brain barrier crossing; enhancing plasma membrane rupture during cell death, leading to the release of inflammatory mediators and further tissue damage. This review explores the emerging evidence for NINJ1's involvement in MS. It discusses how NINJ1 might mediate the migration of immune cells across the blood-brain barrier, exacerbate neuroinflammation, and participate in plasma membrane rupture-related damage. Finally, the review examines potential therapeutic strategies targeting NINJ1 for improved MS management. Abbreviations: MS, Multiple sclerosis; CNS, Central nervous system; BBB, Blood-brain barrier; GSDMD, Gasdermin-D; EAE, Experimental autoimmune encephalitis; HMGB-1, High mobility group box-1 protein; LDH, Lactate dehydrogenase; PMR, Plasma membrane rupture; DMF, Dimethyl fumarate; DUSP1, Dual-specificity phosphatase 1; PAMPs, Pathogen-associated molecular patterns; DAMPs, Danger-associated molecular patterns; PRRs, Pattern recognition receptors; GM-CSF, Granulocyte-macrophage colony stimulating factor; IFN-γ, Interferon gamma; TNF, Tumor necrosis factor; APCs, Antigen-presenting cells; ECs, Endothelial cells; TGF-β, Transforming growth factor-β; PBMCs, Peripheral blood mononuclear cells; FACS, Fluorescence-activated cell sorting; MCP-1, Monocyte chemoattractant protein-1; NLRP3, Pyrin domain-containing 3; TCR, T cell receptor; ROS, Reactive oxygen species; AP-1, Activator protein-1; ANG1, Angiopoietin 1; BMDMs, Bone marrow-derived macrophages; Arp2/3, actin-related protein 2/3; EMT, epithelial-mesenchymal transition; FAK, focal adhesion kinase; LIMK1, LIM domain kinase 1; PAK1, p21-activated kinases 1; Rac1, Ras-related C3 botulinum toxin substrate 1; β-cat, β-caten; MyD88, myeloid differentiation primary response gene 88; TIRAP, Toll/interleukin-1 receptor domain-containing adapter protein; TLR4, Toll-like receptor 4; IRAKs, interleukin-1 receptor-associated kinases; TRAF6, TNF receptor associated factor 6; TAB2/3, TAK1 binding protein 2/3; TAK1, transforming growth factor-β-activated kinase 1; JNK, c-Jun N-terminal kinase; ERK1/2, Extracellular Signal Regulated Kinase 1/2; IKK, inhibitor of kappa B kinase; IκB, inhibitor of NF-κB; NF-κB, nuclear factor kappa-B; AP-1, activator protein-1; ASC, Apoptosis-associated Speck-like protein containing a CARD; NEK7, NIMA-related kinase 7; NLRP3, Pyrin domain-containing 3; CREB, cAMP response element-binding protein.
Collapse
Affiliation(s)
- Yinbin Xu
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Enhao Zhang
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Liangzhe Wei
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Zifeng Dai
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Siqi Chen
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China
| | - Shengjun Zhou
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China.
| | - Yi Huang
- Department of Neurosurgery, Ningbo Key Laboratory of Nervous System and Brain Function, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315010, China; Key Laboratory of Precision Medicine for Atherosclerotic Diseases of Zhejiang Province, Ningbo, Zhejiang 315010, China.
| |
Collapse
|
7
|
Lyubomudrov M, Babkina A, Tsokolaeva Z, Yadgarov M, Shigeev S, Sundukov D, Golubev A. Morphology of Cortical Microglia in the Hyperacute Phase of Subarachnoid Hemorrhage. BIOLOGY 2024; 13:917. [PMID: 39596872 PMCID: PMC11591589 DOI: 10.3390/biology13110917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 11/06/2024] [Accepted: 11/09/2024] [Indexed: 11/29/2024]
Abstract
Hemorrhagic stroke is the deadliest type of stroke. Cellular and molecular biomarkers are important for understanding the pathophysiology of stroke. Microglia are among the most promising biological markers. However, the morphological and physiological characteristics of microglia, as well as the structural and functional aspects of their interactions with neurons and other cells, are largely unknown. Due to the large number of different morphological phenotypes and very limited information on microglial changes in subarachnoid hemorrhage (SAH), we performed this study aimed at identifying the features of the distribution of various microglial phenotypes in the layers of the cerebral cortex in the hyperacute phase of non-traumatic SAH. We studied the distribution of various microglial phenotypes in the layers of the cerebral cortex of SAH non-survivors with a control group (coronary heart disease and sudden cardiac death were the underlying causes of death). An immunohistochemical study using antibodies to iba-1 (a marker of microglia) revealed changes in the morphological phenotypes of microglia in the cerebral cortex after subarachnoid hemorrhage. Significant differences between the groups indicate a rapid microglial response to injury. The findings indicate that there are quantitative and phenotypic changes in microglia in the cerebral cortex during early SAH in the human cortex.
Collapse
Affiliation(s)
- Maksim Lyubomudrov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Anastasiya Babkina
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Zoya Tsokolaeva
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Mikhail Yadgarov
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
| | - Sergey Shigeev
- Bureau of Forensic Medical Examination of the Department of Healthcare of the City of Moscow, Moscow 115516, Russia
| | - Dmitriy Sundukov
- Institute of Medicine, Peoples’ Friendship University of Russia Named after Patrice Lumumba, Moscow 117198, Russia
| | - Arkady Golubev
- Federal Research and Clinical Center of Intensive Care Medicine and Rehabilitology, Moscow 107031, Russia
- Institute of Medicine, Peoples’ Friendship University of Russia Named after Patrice Lumumba, Moscow 117198, Russia
| |
Collapse
|
8
|
Awad M, Sayed RKA, Mohammadin D, Hussein MM, Mokhtar DM. Structural characteristics and regenerative potential: Insights from the molly fish spinal cord. Microsc Res Tech 2024; 87:2643-2653. [PMID: 38923674 DOI: 10.1002/jemt.24633] [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: 01/16/2024] [Revised: 05/16/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024]
Abstract
Unlike mammals, species such as fish and amphibians can regenerate damaged spinal cords, offering insights into potential therapeutic targets. This study investigates the structural features of the molly fish spinal cord through light and electron microscopy. The most notable characteristic was the presence of Mauthner cells (M-cells), which exhibited large cell bodies and processes, as well as synaptic connections with astrocytes. These astrocytic connections contained synaptic vesicles, suggesting electrical transmission at the M-cell endings. Astrocytes, which were labeled with glial fibrillary acidic protein (GFAP), contained cytoplasmic glycogen granules, potentially serving as an emergency fuel source. Two types of oligodendrocytes were identified: a small, dark cell and a larger, lighter cell, both of which reacted strongly with oligodendrocyte transcription factor 2 (Olig2). The dark oligodendrocyte resembled human oligodendrocyte precursors, while the light oligodendrocyte was similar to mature human oligodendrocytes. Additionally, proliferative neurons in the substantia grisea centralis expressed myostatin, Nrf2, and Sox9. Collectively, these findings suggest that the molly fish spinal cord has advanced structural features conducive to spinal cord regeneration and could serve as an excellent model for studying central nervous system regeneration. Further studies on the functional aspects of the molly fish spinal cord are recommended. RESEARCH HIGHLIGHTS: Mauthner cells (M-cell), with their typical large cell body and processes, were the most characteristic feature in Molly fish spinal cord, where it presented synaptic connections with astrocytes and their ends contained synaptic vesicles indicating an electrical transmission in the M-cells endings. Two types of oligodendrocytes could be recognized; both reacted intensely with Oligodendrocyte transcription factor 2 (Olig2). The proliferative neurons of the substantia grisea centralis expressed myostatin, Nrf2, and Sox9. The findings of this study suggest that molly fish possess highly developed structural features conducive to spinal cord regeneration. Consequently, they could be deemed an exemplary model for investigating central nervous system regeneration.
Collapse
Affiliation(s)
- Mahmoud Awad
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Ramy K A Sayed
- Department of Histology, Faculty of Veterinary Medicine, Sohag University, Sohag, Egypt
| | - Dalia Mohammadin
- Department of Histology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Marwa M Hussein
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Doaa M Mokhtar
- Department of Cell and Tissues, Faculty of Veterinary Medicine, Assiut University, Assiut, Egypt
- Department of Histology and Anatomy, School of Veterinary Medicine, Badr University in Assiut, Assiut, Egypt
| |
Collapse
|
9
|
Pang B, Wu L, Peng Y. In vitro modelling of the neurovascular unit for ischemic stroke research: Emphasis on human cell applications and 3D model design. Exp Neurol 2024; 381:114942. [PMID: 39222766 DOI: 10.1016/j.expneurol.2024.114942] [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: 03/15/2024] [Revised: 07/16/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Ischemic stroke has garnered global medical attention as one of the most serious cerebrovascular diseases. The mechanisms involved in both the development and recovery phases of ischemic stroke are complex, involving intricate interactions among different types of cells, each with its own unique functions. To better understand the possible pathogenesis, neurovascular unit (NVU), a concept comprising neurons, endothelial cells, mural cells, glial cells, and extracellular matrix components, has been used in analysing various brain diseases, particularly in ischemic stroke, aiming to depict the interactions between cerebral vasculature and neural cells. While in vivo models often face limitations in terms of reproducibility and the ability to precisely mimic human pathophysiology, it is now important to establish in vitro NVU models for ischemic stroke research. In order to accurately portray the pathological processes occurring within the brain, a diverse array of NVU 2D and 3D in vitro models, each possessing unique characteristics and advantages, have been meticulously developed. This review presents a comprehensive overview of recent advancements in in vitro models specifically tailored for investigating ischemic stroke. Through a systematic categorization of these developments, we elucidate the intricate links between NVU components and the pathogenesis of ischemic stroke. Furthermore, we explore the distinct advantages offered by innovative NVU models, notably 3D models, which closely emulate in vivo conditions. Additionally, an examination of current therapeutic modalities for ischemic stroke developed utilizing in vitro NVU models is provided. Serving as a valuable reference, this review aids in the design and implementation of effective in vitro models for ischemic stroke research.
Collapse
Affiliation(s)
- Bo Pang
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Lei Wu
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China
| | - Ying Peng
- State Key Laboratory of Bioactive Substances and Functions of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100050, China.
| |
Collapse
|
10
|
Wright B, King S, Suphioglu C. The Importance of Phosphoinositide 3-Kinase in Neuroinflammation. Int J Mol Sci 2024; 25:11638. [PMID: 39519189 PMCID: PMC11546674 DOI: 10.3390/ijms252111638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 10/23/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Neuroinflammation, characterised by the activation of immune cells in the central nervous system (CNS), plays a dual role in both protecting against and contributing to the progression of neurodegenerative diseases, such as Alzheimer's disease (AD) and multiple sclerosis (MS). This review explores the role of phosphoinositide 3-kinase (PI3K), a key enzyme involved in cellular survival, proliferation, and inflammatory responses, within the context of neuroinflammation. Two PI3K isoforms of interest, PI3Kγ and PI3Kδ, are specific to the regulation of CNS cells, such as microglia, astrocytes, neurons, and oligodendrocytes, influencing pathways, such as Akt, mTOR, and NF-κB, that control cytokine production, immune cell activation, and neuroprotection. The dysregulation of PI3K signalling is implicated in chronic neuroinflammation, contributing to the exacerbation of neurodegenerative diseases. Preclinical studies show promise in targeting neuronal disorders using PI3K inhibitors, such as AS605240 (PI3Kγ) and idelalisib (PI3Kδ), which have reduced inflammation, microglial activation, and neuronal death in in vivo models of AD. However, the clinical translation of these inhibitors faces challenges, including blood-brain barrier (BBB) permeability, isoform specificity, and long-term safety concerns. This review highlights the therapeutic potential of PI3K modulation in neuroinflammatory diseases, identifying key gaps in the current research, particularly in the need for brain-penetrating and isoform-specific inhibitors. These findings underscore the importance of future research to develop targeted therapies that can effectively modulate PI3K activity and provide neuroprotection in chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- Brock Wright
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| | - Samuel King
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| | - Cenk Suphioglu
- NeuroAllergy Research Laboratory (NARL), School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia; (B.W.); (S.K.)
- Centre for Sustainable Bioproducts, School of Life and Environmental Sciences, Faculty of Science, Engineering and Built Environment, Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, 75 Pigdons Road, Geelong, VIC 3216, Australia
| |
Collapse
|
11
|
das Neves SP, Delivanoglou N, Ren Y, Cucuzza CS, Makuch M, Almeida F, Sanchez G, Barber MJ, Rego S, Schrader R, Faroqi AH, Thomas JL, McLean PJ, Oliveira TG, Irani SR, Piehl F, Da Mesquita S. Meningeal lymphatic function promotes oligodendrocyte survival and brain myelination. Immunity 2024; 57:2328-2343.e8. [PMID: 39217987 PMCID: PMC11464205 DOI: 10.1016/j.immuni.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 04/17/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024]
Abstract
The precise neurophysiological changes prompted by meningeal lymphatic dysfunction remain unclear. Here, we showed that inducing meningeal lymphatic vessel ablation in adult mice led to gene expression changes in glial cells, followed by reductions in mature oligodendrocyte numbers and specific lipid species in the brain. These phenomena were accompanied by altered meningeal adaptive immunity and brain myeloid cell activation. During brain remyelination, meningeal lymphatic dysfunction provoked a state of immunosuppression that contributed to delayed spontaneous oligodendrocyte replenishment and axonal loss. The deficiencies in mature oligodendrocytes and neuroinflammation due to impaired meningeal lymphatic function were solely recapitulated in immunocompetent mice. Patients diagnosed with multiple sclerosis presented reduced vascular endothelial growth factor C in the cerebrospinal fluid, particularly shortly after clinical relapses, possibly indicative of poor meningeal lymphatic function. These data demonstrate that meningeal lymphatics regulate oligodendrocyte function and brain myelination, which might have implications for human demyelinating diseases.
Collapse
Affiliation(s)
- Sofia P das Neves
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Chiara Starvaggi Cucuzza
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Centre for Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Mateusz Makuch
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK
| | - Francisco Almeida
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Guadalupe Sanchez
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Megan J Barber
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Shanon Rego
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Post-baccalaureate Research Education Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Racquelle Schrader
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Post-baccalaureate Research Education Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Ayman H Faroqi
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Jean-Leon Thomas
- Department of Neurology, Yale University School of Medicine, New Haven, CT, USA; Paris Brain Institute, Université Pierre et Marie Curie Paris 06 UMRS1127, Sorbonne Université, Paris Brain Institute, Paris, France
| | - Pamela J McLean
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Tiago Gil Oliveira
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus Gualtar, 4710-057 Braga, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães, Portugal; Department of Neuroradiology, Hospital de Braga, 4710-243 Braga, Portugal
| | - Sarosh R Irani
- Oxford Autoimmune Neurology Group, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Department of Clinical Neurology, John Radcliffe Hospital, Oxford, UK
| | - Fredrik Piehl
- Department of Clinical Neuroscience, Karolinska Institutet, Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden; Centre for Neurology, Academic Specialist Center, Stockholm Health Services, Stockholm, Sweden
| | - Sandro Da Mesquita
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA; Neuroscience Ph.D. Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Jacksonville, FL 32224, USA.
| |
Collapse
|
12
|
Fanlo-Ucar H, Picón-Pagès P, Herrera-Fernández V, ILL-Raga G, Muñoz FJ. The Dual Role of Amyloid Beta-Peptide in Oxidative Stress and Inflammation: Unveiling Their Connections in Alzheimer's Disease Etiopathology. Antioxidants (Basel) 2024; 13:1208. [PMID: 39456461 PMCID: PMC11505517 DOI: 10.3390/antiox13101208] [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: 08/07/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease, and it is currently the seventh leading cause of death worldwide. It is characterized by the extracellular aggregation of the amyloid β-peptide (Aβ) into oligomers and fibrils that cause synaptotoxicity and neuronal death. Aβ exhibits a dual role in promoting oxidative stress and inflammation. This review aims to unravel the intricate connection between these processes and their contribution to AD progression. The review delves into oxidative stress in AD, focusing on the involvement of metals, mitochondrial dysfunction, and biomolecule oxidation. The distinct yet overlapping concept of nitro-oxidative stress is also discussed, detailing the roles of nitric oxide, mitochondrial perturbations, and their cumulative impact on Aβ production and neurotoxicity. Inflammation is examined through astroglia and microglia function, elucidating their response to Aβ and their contribution to oxidative stress within the AD brain. The blood-brain barrier and oligodendrocytes are also considered in the context of AD pathophysiology. We also review current diagnostic methodologies and emerging therapeutic strategies aimed at mitigating oxidative stress and inflammation, thereby offering potential treatments for halting or slowing AD progression. This comprehensive synthesis underscores the pivotal role of Aβ in bridging oxidative stress and inflammation, advancing our understanding of AD and informing future research and treatment paradigms.
Collapse
Affiliation(s)
- Hugo Fanlo-Ucar
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Pol Picón-Pagès
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
- Laboratory of Molecular and Cellular Neurobiotechnology, Institute of Bioengineering of Catalonia (IBEC), 08028 Barcelona, Spain
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), 08028 Barcelona, Spain
| | - Víctor Herrera-Fernández
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Gerard ILL-Raga
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| | - Francisco J. Muñoz
- Laboratory of Molecular Physiology, Department of Medicine and Life Sciences, Faculty of Medicine and Life Sciences, Universitat Pompeu Fabra, 08003 Barcelona, Spain; (H.F.-U.); (P.P.-P.); (V.H.-F.); (G.I.-R.)
| |
Collapse
|
13
|
Tang W, Wang Q, Sun M, Liu C, Huang Y, Zhou M, Zhang X, Meng Z, Zhang J. The gut microbiota-oligodendrocyte axis: A promising pathway for modulating oligodendrocyte homeostasis and demyelination-associated disorders. Life Sci 2024; 354:122952. [PMID: 39127317 DOI: 10.1016/j.lfs.2024.122952] [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: 04/23/2024] [Revised: 07/22/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
The bidirectional regulation between the gut microbiota and brain, known as gut-brain axis, has received significant attention. The myelin sheath, produced by oligodendrocytes or Schwann cells, is essential for efficient nervous signal transmission and the maintenance of brain function. Growing evidence shows that both oligodendrogenesis and myelination are modulated by gut microbiota and its metabolites, and when dysbiosis occurs, changes in the microbiota composition and/or associated metabolites may impact developmental myelination and the occurrence of neurodevelopmental disabilities. Although the link between the microbiota and demyelinating disease such as multiple sclerosis has been extensively studied, our knowledge about the role of the microbiota in other myelin-related disorders, such as neurodegenerative diseases, is limited. Mechanistically, the microbiota-oligodendrocyte axis is primarily mediated by factors such as inflammation, the vagus nerve, endocrine hormones, and microbiota metabolites as evidenced by metagenomics, metabolomics, vagotomy, and morphological and molecular approaches. Treatments targeting this axis include probiotics, prebiotics, microbial metabolites, herbal bioactive compounds, and specific dietary management. In addition to the commonly used approaches, viral vector-mediated tracing and gene manipulation, integrated multiomics and multicenter clinical trials will greatly promote the mechanistic and interventional studies and ultimately, the development of new preventive and therapeutic strategies against gut-oligodendrocyte axis-mediated brain impairments. Interestingly, recent findings showed that microbiota dysbiosis can be induced by hippocampal myelin damage and is reversible by myelin-targeted drugs, which provides new insights into understanding how hippocampus-based functional impairment (such as in neurodegenerative Alzheimer's disease) regulates the peripheral homeostasis of microbiota and associated systemic disorders.
Collapse
Affiliation(s)
- Wen Tang
- Department of Gastroenterology, Chongqing Western Hospital, Chongqing 400052, China
| | - Qi Wang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Mingguang Sun
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China; Department of Neurology, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, Beijing University of Chinese Medicine, Beijing 100853, China
| | - Chang''e Liu
- Department of Nutrition, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Yonghua Huang
- Department of Neurology, The Seventh Medical Center of Chinese PLA General Hospital, Beijing 100700, China
| | - Maohu Zhou
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Xuan Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China
| | - Zhaoyou Meng
- Department of Neurology, Xinqiao Hospital, Army Medical University, Chongqing 400037, China.
| | - Jiqiang Zhang
- Department of Neurobiology, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
14
|
Theophanous S, Sargiannidou I, Kleopa KA. Glial Cells as Key Regulators in Neuroinflammatory Mechanisms Associated with Multiple Sclerosis. Int J Mol Sci 2024; 25:9588. [PMID: 39273535 PMCID: PMC11395575 DOI: 10.3390/ijms25179588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
Even though several highly effective treatments have been developed for multiple sclerosis (MS), the underlying pathological mechanisms and drivers of the disease have not been fully elucidated. In recent years, there has been a growing interest in studying neuroinflammation in the context of glial cell involvement as there is increasing evidence of their central role in disease progression. Although glial cell communication and proper function underlies brain homeostasis and maintenance, their multiple effects in an MS brain remain complex and controversial. In this review, we aim to provide an overview of the contribution of glial cells, oligodendrocytes, astrocytes, and microglia in the pathology of MS during both the activation and orchestration of inflammatory mechanisms, as well as of their synergistic effects during the repair and restoration of function. Additionally, we discuss how the understanding of glial cell involvement in MS may provide new therapeutic targets either to limit disease progression or to facilitate repair.
Collapse
Affiliation(s)
- Styliani Theophanous
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| | - Kleopas A Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics, 2371 Nicosia, Cyprus
| |
Collapse
|
15
|
Shen Y, Liu F, Zhang M. Therapeutic potential of plant-derived natural compounds in Alzheimer's disease: Targeting microglia-mediated neuroinflammation. Biomed Pharmacother 2024; 178:117235. [PMID: 39094545 DOI: 10.1016/j.biopha.2024.117235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/27/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
Microglia are resident immune cells of the central nervous system (CNS) with roles in sensing, housekeeping, and defense. Exploring the role of microglia in the occurrence and development of Alzheimer's disease (AD) and the possible therapeutic mechanism of plant-derived natural compounds (PDNCs) that regulate microglia-associated neuroinflammation may potentially help in elucidating the pathogenesis of AD and provide novel insights for its treatment. This review explores the role of abnormal microglial activation and its dominant neuroinflammatory response, as well as the activation of their target receptors and signaling pathways in AD pathogenesis. Additionally, we report an update on the potential pharmacological mechanisms of multiple PDNCs in modulating microglia-associated neuroinflammation in AD treatment. Dysregulated activation of microglial receptors and their downstream pathways impaired immune homeostasis in animal models of AD. Multiple signaling pathways, such as mitogen-activated protein kinase (MAPK), nuclear factor kappa light chain enhancer of activated B cells (NF-κB), and Toll-like receptors, play important roles in microglial activation and can exacerbate microglia-mediated neuroinflammation. PDNCs, such as magnolol, stigmasterol, matrine, naringenin, naringin, and resveratrol, can delay the progression of AD by inhibiting the proinflammatory receptors of microglia, activating its anti-inflammatory receptors, regulating the receptors related to β-amyloid (Aβ) clearance, reversing immune dysregulation, and maintaining the immune homeostasis of microglial downstream pathways. This review summarizes the mechanisms by which microglia cause chronic inflammation in AD and evaluates the beneficial effects of PDNCs on immune regulation in AD by regulating microglial receptors and their downstream pathways.
Collapse
Affiliation(s)
- Yanyan Shen
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| | - Fang Liu
- Department of Neurology, The First Affiliated Hospital of China Medical University, China
| | - Mingjie Zhang
- Department of Neurosurgery, Shengjing Hospital of China Medical University, China.
| |
Collapse
|
16
|
Mazziotti V, Crescenzo F, Turano E, Guandalini M, Bertolazzo M, Ziccardi S, Virla F, Camera V, Marastoni D, Tamanti A, Calabrese M. The contribution of tumor necrosis factor to multiple sclerosis: a possible role in progression independent of relapse? J Neuroinflammation 2024; 21:209. [PMID: 39169320 PMCID: PMC11340196 DOI: 10.1186/s12974-024-03193-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024] Open
Abstract
Tumor necrosis factor (TNF) is a pleiotropic cytokine regulating many physiological and pathological immune-mediated processes. Specifically, it has been recognized as an essential pro-inflammatory cytokine implicated in multiple sclerosis (MS) pathogenesis and progression. MS is a chronic immune-mediated disease of the central nervous system, characterized by multifocal acute and chronic inflammatory demyelination in white and grey matter, along with neuroaxonal loss. A recent concept in the field of MS research is disability resulting from Progression Independent of Relapse Activity (PIRA). PIRA recognizes that disability accumulation since the early phase of the disease can occur independently of relapse activity overcoming the traditional dualistic view of MS as either a relapsing-inflammatory or a progressive-neurodegenerative disease. Several studies have demonstrated an upregulation in TNF expression in both acute and chronic active MS brain lesions. Additionally, elevated TNF levels have been observed in the serum and cerebrospinal fluid of MS patients. TNF appears to play a significant role in maintaining chronic intrathecal inflammation, promoting axonal damage neurodegeneration, and consequently contributing to disease progression and disability accumulation. In summary, this review highlights the current understanding of TNF and its receptors in MS progression, specifically focusing on the relatively unexplored PIRA condition. Further research in this area holds promise for potential therapeutic interventions targeting TNF to mitigate disability in MS patients.
Collapse
Affiliation(s)
- Valentina Mazziotti
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Francesco Crescenzo
- Neurology Unit - Multiple Sclerosis Center, Scaligera Local Unit of Health and Social Services 9, Mater Salutis Hospital, 37045, Legnago, Verona, Italy
| | - Ermanna Turano
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Maddalena Guandalini
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Maddalena Bertolazzo
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Stefano Ziccardi
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Federica Virla
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Valentina Camera
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Damiano Marastoni
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Agnese Tamanti
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy
| | - Massimiliano Calabrese
- Neurology B Unit - Multiple Sclerosis Center, Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37134, Verona, Italy.
| |
Collapse
|
17
|
Ma W, Geng Y, Liu Y, Pan H, Wang Q, Zhang Y, Wang L. The mechanisms of white matter injury and immune system crosstalk in promoting the progression of Parkinson's disease: a narrative review. Front Aging Neurosci 2024; 16:1345918. [PMID: 38863783 PMCID: PMC11165104 DOI: 10.3389/fnagi.2024.1345918] [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: 11/29/2023] [Accepted: 05/15/2024] [Indexed: 06/13/2024] Open
Abstract
Parkinson's disease (PD) is neurodegenerative disease in middle-aged and elderly people with some pathological mechanisms including immune disorder, neuroinflammation, white matter injury and abnormal aggregation of alpha-synuclein, etc. New research suggests that white matter injury may be important in the development of PD, but how inflammation, the immune system, and white matter damage interact to harm dopamine neurons is not yet understood. Therefore, it is particularly important to delve into the crosstalk between immune cells in the central and peripheral nervous system based on the study of white matter damage in PD. This crosstalk could not only exacerbate the pathological process of PD but may also reveal new therapeutic targets. By understanding how immune cells penetrate through the blood-brain barrier and activate inflammatory responses within the central nervous system, we can better grasp the impact of structural destruction of white matter in PD and explore how this process can be modulated to mitigate or combat disease progression. Microglia, astrocytes, oligodendrocytes and peripheral immune cells (especially T cells) play a central role in its pathological process where these immune cells produce and respond to pro-inflammatory cytokines such as tumor necrosis factor (TNF-α), interleukin-1β(IL-1β) and interleukin-6(IL-6), and white matter injury causes microglia to become pro-inflammatory and release inflammatory mediators, which attract more immune cells to the damaged area, increasing the inflammatory response. Moreover, white matter damage also causes dysfunction of blood-brain barrier, allows peripheral immune cells and inflammatory factors to invade the brain further, and enhances microglia activation forming a vicious circle that intensifies neuroinflammation. And these factors collectively promote the neuroinflammatory environment and neurodegeneration changes of PD. Overall, these findings not only deepen our understanding of the complexity of PD, but also provide new targets for the development of therapeutic strategies focused on inflammation and immune regulation mechanisms. In summary, this review provided the theoretical basis for clarifying the pathogenesis of PD, summarized the association between white matter damage and the immune cells in the central and peripheral nervous systems, and then emphasized their potential specific mechanisms of achieving crosstalk with further aggravating the pathological process of PD.
Collapse
Affiliation(s)
- Wen Ma
- Graduate School of Education, Shandong Sport University, Jinan, Shandong, China
| | - Yifan Geng
- Xuzhou Clinical School, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Youhan Liu
- Graduate School of Education, Shandong Sport University, Jinan, Shandong, China
| | - Huixin Pan
- Graduate School of Education, Shandong Sport University, Jinan, Shandong, China
| | - Qinglu Wang
- Graduate School of Education, Shandong Sport University, Jinan, Shandong, China
| | - Yaohua Zhang
- Key Laboratory of Biomedical Engineering & Technology of Shandong High School, Qilu Medical University, Zibo, China
| | - Liping Wang
- Graduate School of Education, Shandong Sport University, Jinan, Shandong, China
| |
Collapse
|
18
|
Wu Y, Libby JB, Dumitrescu L, De Jager PL, Menon V, Schneider JA, Bennett DA, Hohman TJ. Association of 10 VEGF Family Genes with Alzheimer's Disease Endophenotypes at Single Cell Resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589221. [PMID: 38826287 PMCID: PMC11142115 DOI: 10.1101/2024.04.12.589221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The cell-type specific role of the vascular endothelial growth factors (VEGFs) in the pathogenesis of Alzheimer's disease (AD) is not well characterized. In this study, we utilized a single-nucleus RNA sequencing dataset from Dorsolateral Prefrontal Cortex (DLFPC) of 424 donors from the Religious Orders Study and Memory and Aging Project (ROS/MAP) to investigate the effect of 10 VEGF genes ( VEGFA, VEGFB, VEGFC, VEGFD, PGF, FLT1, FLT4, KDR, NRP1 , and NRP2 ) on AD endophenotypes. Mean age of death was 89 years, among which 68% were females, and 52% has AD dementia. Negative binomial mixed models were used for differential expression analysis and for association analysis with β-amyloid load, PHF tau tangle density, and both cross-sectional and longitudinal global cognitive function. Intercellular VEGF-associated signaling was profiled using CellChat. We discovered prefrontal cortical FLT1 expression was upregulated in AD brains in both endothelial and microglial cells. Higher FLT1 expression was also associated with worse cross-sectional global cognitive function, longitudinal cognitive trajectories, and β-amyloid load. Similarly, higher endothelial FLT4 expression was associated with more β-amyloid load. In contrast to the receptors, VEGFB showed opposing effects on β-amyloid load whereby higher levels in oligodendrocytes was associated with high amyloid burden, while higher levels in inhibitory neurons was associated with lower amyloid burden. Finally, AD cells showed significant reduction in overall VEGF signaling comparing to those from cognitive normal participants. Our results highlight key changes in VEGF receptor expression in endothelial and microglial cells during AD, and the potential protective role of VEGFB in neurons.
Collapse
|
19
|
Schreiner TG, Schreiner OD, Ciobanu RC. Spinal Cord Injury Management Based on Microglia-Targeting Therapies. J Clin Med 2024; 13:2773. [PMID: 38792314 PMCID: PMC11122315 DOI: 10.3390/jcm13102773] [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: 04/19/2024] [Revised: 05/05/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
Spinal cord injury is a complicated medical condition both from the clinician's point of view in terms of management and from the patient's perspective in terms of unsatisfactory recovery. Depending on the severity, this disorder can be devastating despite the rapid and appropriate use of modern imaging techniques and convenient surgical spinal cord decompression and stabilization. In this context, there is a mandatory need for novel adjunctive therapeutic approaches to classical treatments to improve rehabilitation chances and clinical outcomes. This review offers a new and original perspective on therapies targeting the microglia, one of the most relevant immune cells implicated in spinal cord disorders. The first part of the manuscript reviews the anatomical and pathophysiological importance of the blood-spinal cord barrier components, including the role of microglia in post-acute neuroinflammation. Subsequently, the authors present the emerging therapies based on microglia modulation, such as cytokines modulators, stem cell, microRNA, and nanoparticle-based treatments that could positively impact spinal cord injury management. Finally, future perspectives and challenges are also highlighted based on the ongoing clinical trials related to medications targeting microglia.
Collapse
Affiliation(s)
- Thomas Gabriel Schreiner
- Department of Medical Specialties III, Faculty of Medicine, University of Medicine and Pharmacy “Grigore T. Popa”, 700115 Iasi, Romania;
- First Neurology Clinic, “Prof. Dr. N. Oblu” Clinical Emergency Hospital, 700309 Iasi, Romania
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 700050 Iasi, Romania;
| | - Oliver Daniel Schreiner
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 700050 Iasi, Romania;
- Medical Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Romeo Cristian Ciobanu
- Department of Electrical Measurements and Materials, Faculty of Electrical Engineering and Information Technology, Gheorghe Asachi Technical University of Iasi, 700050 Iasi, Romania;
| |
Collapse
|
20
|
Li Y, Li P, Tao Q, Abuqeis IJA, Xiyang Y. Role and limitation of cell therapy in treating neurological diseases. IBRAIN 2024; 10:93-105. [PMID: 38682022 PMCID: PMC11045202 DOI: 10.1002/ibra.12152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 05/01/2024]
Abstract
The central role of the brain in governing systemic functions within human physiology underscores its paramount significance as the focal point of physiological regulation. The brain, a highly sophisticated organ, orchestrates a diverse array of physiological processes encompassing motor control, sensory perception, cognition, emotion, and the regulation of vital functions, such as heartbeat, respiration, and hormonal equilibrium. A notable attribute of neurological diseases manifests as the depletion of neurons and the occurrence of tissue necrosis subsequent to injury. The transplantation of neural stem cells (NSCs) into the brain exhibits the potential for the replacement of lost neurons and the reconstruction of neural circuits. Furthermore, the transplantation of other types of cells in alternative locations can secrete nutritional factors that indirectly contribute to the restoration of nervous system equilibrium and the mitigation of neural inflammation. This review summarized a comprehensive investigation into the role of NSCs, hematopoietic stem cells, mesenchymal stem cells, and support cells like astrocytes and microglia in alleviating neurological deficits after cell infusion. Moreover, a thorough assessment was undertaken to discuss extant constraints in cellular transplantation therapies, concurrently delineating indispensable model-based methodologies, specifically on organoids, which were essential for guiding prospective research initiatives in this specialized field.
Collapse
Affiliation(s)
- Yu‐Qi Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Peng‐Fei Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | - Qian Tao
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational MedicineKunming University of Science and TechnologyKunmingChina
| | | | - Yan‐Bin Xiyang
- School of Basic MedicineKunming Medical UniversityKunmingChina
- Department of Pharmacology and Toxicology, College of PharmacologyUniversity of ArizonaTucsonArizonaUSA
| |
Collapse
|
21
|
Nguyen KL, Bhatt IJ, Gupta S, Showkat N, Swanson KA, Fischer R, Kontermann RE, Pfizenmaier K, Bracchi-Ricard V, Bethea JR. Tumor necrosis factor receptor 2 activation elicits sex-specific effects on cortical myelin proteins and functional recovery in a model of multiple sclerosis. Brain Res Bull 2024; 207:110885. [PMID: 38246200 PMCID: PMC10923072 DOI: 10.1016/j.brainresbull.2024.110885] [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: 09/30/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 01/23/2024]
Abstract
Multiple sclerosis (MS), a demyelinating autoimmune disease of the central nervous system (CNS), predominately affects females compared to males. Tumor necrosis factor (TNF), a pro-inflammatory cytokine, signaling through TNF receptor 1 contributes to inflammatory disease pathogenesis. In contrast, TNF receptor 2 signaling is neuroprotective. Current anti-TNF MS therapies are shown to be detrimental to patients due to pleiotropic effects on both pro- and anti-inflammatory functions. Using a non-pertussis toxin (nPTX) experimental autoimmune encephalomyelitis (EAE) model in C57BL/6 mice, we systemically administered a TNFR2 agonist (p53-sc-mTNFR2) to investigate behavioral and pathophysiological changes in both female and male mice. Our data shows that TNFR2 activation alleviates motor and sensory symptoms in females. However, in males, the agonist only alleviates sensory symptoms and not motor. nPTX EAE induction in TNFR2 global knockout mice caused exacerbated motor symptoms in females along with an earlier day of onset, but not in males. Our data demonstrates that TNFR2 agonist efficacy is sex-specific for alleviation of motor symptoms, however, it effectively reduces mechanical hypersensitivity in both females and males. Altogether, these data support the therapeutic promise TNFR2 agonism holds as an MS therapeutic and, more broadly, to treat central neuropathic pain.
Collapse
MESH Headings
- Humans
- Male
- Female
- Mice
- Animals
- Multiple Sclerosis
- Receptors, Tumor Necrosis Factor, Type II/agonists
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/therapeutic use
- Tumor Necrosis Factor Inhibitors/therapeutic use
- Mice, Inbred C57BL
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Myelin Proteins
- Tumor Necrosis Factor-alpha/metabolism
- Mice, Knockout
Collapse
Affiliation(s)
- Kayla L Nguyen
- Department of Anatomy and Cell Biology, The George Washington University, Washington, DC 20052, United States.
| | - Ishaan J Bhatt
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Shruti Gupta
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Nazaf Showkat
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Kathryn A Swanson
- Department of Biology, Drexel University, Philadelphia, PA 19104, United States
| | - Roman Fischer
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany; Stuttgart Research Center Systems Biology, University of Stuttgart, 70569 Stuttgart, Germany
| | | | - John R Bethea
- Department of Anatomy and Cell Biology, The George Washington University, Washington, DC 20052, United States.
| |
Collapse
|
22
|
Filannino FM, Panaro MA, Benameur T, Pizzolorusso I, Porro C. Extracellular Vesicles in the Central Nervous System: A Novel Mechanism of Neuronal Cell Communication. Int J Mol Sci 2024; 25:1629. [PMID: 38338906 PMCID: PMC10855168 DOI: 10.3390/ijms25031629] [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: 12/21/2023] [Revised: 01/21/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Cell-to-cell communication is essential for the appropriate development and maintenance of homeostatic conditions in the central nervous system. Extracellular vesicles have recently come to the forefront of neuroscience as novel vehicles for the transfer of complex signals between neuronal cells. Extracellular vesicles are membrane-bound carriers packed with proteins, metabolites, and nucleic acids (including DNA, mRNA, and microRNAs) that contain the elements present in the cell they originate from. Since their discovery, extracellular vesicles have been studied extensively and have opened up new understanding of cell-cell communication; they may cross the blood-brain barrier in a bidirectional way from the bloodstream to the brain parenchyma and vice versa, and play a key role in brain-periphery communication in physiology as well as pathology. Neurons and glial cells in the central nervous system release extracellular vesicles to the interstitial fluid of the brain and spinal cord parenchyma. Extracellular vesicles contain proteins, nucleic acids, lipids, carbohydrates, and primary and secondary metabolites. that can be taken up by and modulate the behaviour of neighbouring recipient cells. The functions of extracellular vesicles have been extensively studied in the context of neurodegenerative diseases. The purpose of this review is to analyse the role extracellular vesicles extracellular vesicles in central nervous system cell communication, with particular emphasis on the contribution of extracellular vesicles from different central nervous system cell types in maintaining or altering central nervous system homeostasis.
Collapse
Affiliation(s)
| | - Maria Antonietta Panaro
- Department of Biosciences, Biotechnologies and Environment, University of Bari, 70125 Bari, Italy;
| | - Tarek Benameur
- Department of Biomedical Sciences, College of Medicine, King Faisal University, Al-Ahsa 31982, Saudi Arabia;
| | - Ilaria Pizzolorusso
- Child and Adolescent Neuropsychiatry Unit, Department of Mental Health, ASL Foggia, 71121 Foggia, Italy;
| | - Chiara Porro
- Department of Clinical and Experimental Medicine, University of Foggia, 71121 Foggia, Italy;
| |
Collapse
|
23
|
Harry GJ. Developmental Associations between Neurovascularization and Microglia Colonization. Int J Mol Sci 2024; 25:1281. [PMID: 38279280 PMCID: PMC10816009 DOI: 10.3390/ijms25021281] [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: 12/30/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
The temporal and spatial pattern of microglia colonization and vascular infiltration of the nervous system implies critical associated roles in early stages of nervous system development. Adding to existing reviews that cover a broad spectrum of the various roles of microglia during brain development, the current review will focus on the developmental ontogeny and interdependency between the colonization of the nervous system with yolk sac derived macrophages and vascularization. Gaining a better understanding of the timing and the interdependency of these two processes will significantly contribute to the interpretation of data generated regarding alterations in either process during early development. Additionally, such knowledge should provide a framework for understanding the influence of the early gestational environmental and the impact of genetics, disease, disorders, or exposures on the early developing nervous system and the potential for long-term and life-time effects.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, Durham, NC 27709, USA
| |
Collapse
|
24
|
Dejbakht M, Akhzari M, Jalili S, Faraji F, Barazesh M. Multiple Sclerosis: New Insights into Molecular Pathogenesis and Novel Platforms for Disease Treatment. Curr Drug Res Rev 2024; 16:175-197. [PMID: 37724675 DOI: 10.2174/2589977516666230915103730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 06/23/2023] [Accepted: 07/03/2023] [Indexed: 09/21/2023]
Abstract
BACKGROUND Multiple sclerosis (MS), a chronic inflammatory disorder, affects the central nervous system via myelin degradation. The cause of MS is not fully known, but during recent years, our knowledge has deepened significantly regarding the different aspects of MS, including etiology, molecular pathophysiology, diagnosis and therapeutic options. Myelin basic protein (MBP) is the main myelin protein that accounts for maintaining the stability of the myelin sheath. Recent evidence has revealed that MBP citrullination or deamination, which is catalyzed by Ca2+ dependent peptidyl arginine deiminase (PAD) enzyme leads to the reduction of positive charge, and subsequently proteolytic cleavage of MBP. The overexpression of PAD2 in the brains of MS patients plays an essential role in new epitope formation and progression of the autoimmune disorder. Some drugs have recently entered phase III clinical trials with promising efficacy and will probably obtain approval in the near future. As different therapeutic platforms develop, finding an optimal treatment for each individual patient will be more challenging. AIMS This review provides a comprehensive insight into MS with a focus on its pathogenesis and recent advances in diagnostic methods and its present and upcoming treatment modalities. CONCLUSION MS therapy alters quickly as research findings and therapeutic options surrounding MS expand. McDonald's guidelines have created different criteria for MS diagnosis. In recent years, ever-growing interest in the development of PAD inhibitors has led to the generation of many reversible and irreversible PAD inhibitors against the disease with satisfactory therapeutic outcomes.
Collapse
Affiliation(s)
- Majid Dejbakht
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Morteza Akhzari
- School of Nursing, Larestan University of Medical Sciences, Larestan, Iran
| | - Sajad Jalili
- Department of Orthopedics, School of Medicine, Ahvaz Jundishapour University of Medical Sciences, Ahvaz, Iran
| | - Fouziyeh Faraji
- Department of Cellular and Molecular Research Center, Gerash University of Medical Sciences, Gerash, Iran
| | - Mahdi Barazesh
- Department of Biotechnology, Cellular and Molecular Research Center, School of Paramedical, Gerash University of Medical Sciences, Gerash, Iran
| |
Collapse
|
25
|
Filho AMC, Gomes NS, Lós DB, Leite IB, Tremblay MÈ, Macêdo DS. Microglia and Microbiome-Gut-Brain Axis. ADVANCES IN NEUROBIOLOGY 2024; 37:303-331. [PMID: 39207699 DOI: 10.1007/978-3-031-55529-9_17] [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
The mammalian gut contains a community of microorganisms called gut microbiome. The gut microbiome is integrated into mammalian physiology, contributing to metabolism, production of metabolites, and promoting immunomodulatory actions. Microglia, the brain's resident innate immune cells, play an essential role in homeostatic neurogenesis, synaptic remodeling, and glial maturation. Microglial dysfunction has been implicated in the pathogenesis of several neuropsychiatric disorders. Recent findings indicate that microglia are influenced by the gut microbiome and their derived metabolites throughout life. The pathways by which microbiota regulate microglia have only started to be understood, but this discovery has the potential to provide valuable insights into the pathogenesis of brain disorders associated with an altered microbiome. Here, we discuss the recent literature on the role of the gut microbiome in modulating microglia during development and adulthood and summarize the key findings on this bidirectional crosstalk in selected examples of neuropsychiatric and neurodegenerative disorders. We also highlight some current caveats and perspectives for the field.
Collapse
Affiliation(s)
- Adriano Maia Chaves Filho
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Nayana Soares Gomes
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Deniele Bezerra Lós
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Isabel Bessa Leite
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada.
- Department of Molecular Medicine, Université de Laval, Québec City, Canada.
- Department of Neurology and Neurosurgery, McGill University, Montréal, Canada.
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Danielle S Macêdo
- Department of Physiology and Pharmacology, Drug Research and Development Center, Faculty of Medicine, Federal University of Ceara, Fortaleza, CE, Brazil.
- National Institute for Translational Medicine (INCT-TM, CNPq), Ribeirão Preto, SP, Brazil.
| |
Collapse
|
26
|
Harry GJ. Microglia Colonization Associated with Angiogenesis and Neural Cell Development. ADVANCES IN NEUROBIOLOGY 2024; 37:163-178. [PMID: 39207692 DOI: 10.1007/978-3-031-55529-9_10] [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
The temporal and spatial pattern of microglia colonization of the nervous system implies a role in early stages of organ development including cell proliferation, differentiation, and neurovascularization. As microglia colonize and establish within the developing nervous system, they assume a neural-specific identity and contribute to key developmental events. Their association around blood vessels implicates them in development of the vascular system or vice versa. A similar association has been reported for neural cell proliferation and associated phenotypic shifts and for cell fate differentiation to neuronal or glial phenotypes. These processes are accomplished by phagocytic activities, cell-cell contact relationships, and secretion of various factors. This chapter will present data currently available from studies evaluating the dynamic and interactive nature of these processes throughout the progression of nervous system development.
Collapse
Affiliation(s)
- G Jean Harry
- Mechanistic Toxicology Branch, Division of Translational Toxicology, National Institute Environmental Health Sciences, Research Triangle Park, NC, USA.
| |
Collapse
|
27
|
Malvaso A, Gatti A, Negro G, Calatozzolo C, Medici V, Poloni TE. Microglial Senescence and Activation in Healthy Aging and Alzheimer's Disease: Systematic Review and Neuropathological Scoring. Cells 2023; 12:2824. [PMID: 38132144 PMCID: PMC10742050 DOI: 10.3390/cells12242824] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/07/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
The greatest risk factor for neurodegeneration is the aging of the multiple cell types of human CNS, among which microglia are important because they are the "sentinels" of internal and external perturbations and have long lifespans. We aim to emphasize microglial signatures in physiologic brain aging and Alzheimer's disease (AD). A systematic literature search of all published articles about microglial senescence in human healthy aging and AD was performed, searching for PubMed and Scopus online databases. Among 1947 articles screened, a total of 289 articles were assessed for full-text eligibility. Microglial transcriptomic, phenotypic, and neuropathological profiles were analyzed comprising healthy aging and AD. Our review highlights that studies on animal models only partially clarify what happens in humans. Human and mice microglia are hugely heterogeneous. Like a two-sided coin, microglia can be protective or harmful, depending on the context. Brain health depends upon a balance between the actions and reactions of microglia maintaining brain homeostasis in cooperation with other cell types (especially astrocytes and oligodendrocytes). During aging, accumulating oxidative stress and mitochondrial dysfunction weaken microglia leading to dystrophic/senescent, otherwise over-reactive, phenotype-enhancing neurodegenerative phenomena. Microglia are crucial for managing Aβ, pTAU, and damaged synapses, being pivotal in AD pathogenesis.
Collapse
Affiliation(s)
- Antonio Malvaso
- IRCCS “C. Mondino” Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.M.); (A.G.)
| | - Alberto Gatti
- IRCCS “C. Mondino” Foundation, National Neurological Institute, Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy; (A.M.); (A.G.)
| | - Giulia Negro
- Department of Neurology, University of Milano Bicocca, 20126 Milan, Italy;
| | - Chiara Calatozzolo
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Abbiategrasso, 20081 Milan, Italy;
| | - Valentina Medici
- Department of Translational Medicine, University of Eastern Piedmont, 28100 Novara, Italy;
| | - Tino Emanuele Poloni
- Department of Neurology and Neuropathology, Golgi-Cenci Foundation, Abbiategrasso, 20081 Milan, Italy;
| |
Collapse
|
28
|
Kaffe D, Kaplanis SI, Karagogeos D. The Roles of Caloric Restriction Mimetics in Central Nervous System Demyelination and Remyelination. Curr Issues Mol Biol 2023; 45:9526-9548. [PMID: 38132442 PMCID: PMC10742427 DOI: 10.3390/cimb45120596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 11/16/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The dysfunction of myelinating glial cells, the oligodendrocytes, within the central nervous system (CNS) can result in the disruption of myelin, the lipid-rich multi-layered membrane structure that surrounds most vertebrate axons. This leads to axonal degeneration and motor/cognitive impairments. In response to demyelination in the CNS, the formation of new myelin sheaths occurs through the homeostatic process of remyelination, facilitated by the differentiation of newly formed oligodendrocytes. Apart from oligodendrocytes, the two other main glial cell types of the CNS, microglia and astrocytes, play a pivotal role in remyelination. Following a demyelination insult, microglia can phagocytose myelin debris, thus permitting remyelination, while the developing neuroinflammation in the demyelinated region triggers the activation of astrocytes. Modulating the profile of glial cells can enhance the likelihood of successful remyelination. In this context, recent studies have implicated autophagy as a pivotal pathway in glial cells, playing a significant role in both their maturation and the maintenance of myelin. In this Review, we examine the role of substances capable of modulating the autophagic machinery within the myelinating glial cells of the CNS. Such substances, called caloric restriction mimetics, have been shown to decelerate the aging process by mitigating age-related ailments, with their mechanisms of action intricately linked to the induction of autophagic processes.
Collapse
Affiliation(s)
- Despoina Kaffe
- Department of Biology, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
| | - Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Vassilika Vouton, 70013 Heraklion, Greece;
- Institute of Molecular Biology & Biotechnology (IMBB), Foundation for Research and Technology-Hellas (FORTH), Vassilika Vouton, 70013 Heraklion, Greece
| |
Collapse
|
29
|
Christodoulou CC, Onisiforou A, Zanos P, Papanicolaou EZ. Unraveling the transcriptomic signatures of Parkinson's disease and major depression using single-cell and bulk data. Front Aging Neurosci 2023; 15:1273855. [PMID: 38020762 PMCID: PMC10664927 DOI: 10.3389/fnagi.2023.1273855] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Background Motor symptoms are well-characterized in Parkinson's disease (PD). However, non-motor symptoms, such as depression, are commonly observed and can appear up to 10 years before motor features, resulting in one-third of individuals being misdiagnosed with a neuropsychiatric disorder. Thus, identifying diagnostic biomarkers is crucial for accurate PD diagnosis during its prodromal or early stages. Methods We employed an integrative approach, combining single nucleus RNA and bulk mRNA transcriptomics to perform comparative molecular signatures analysis between PD and major depressive disorder (MDD). We examined 39,834 nuclei from PD (GSE202210) and 32,707 nuclei from MDD (GSE144136) in the dorsolateral prefrontal cortex (dlPFC) of Brodmann area 9. Additionally, we analyzed bulk mRNA peripheral blood samples from PD compared to controls (GSE49126, GSE72267), as well as MDD compared to controls (GSE39653). Results Our findings show a higher proportion of astrocytes, and oligodendrocyte cells in the dlPFC of individuals with PD vs. MDD. The excitatory to inhibitory neurons (E/I) ratio analysis indicates that MDD has a ratio close to normal 80/20, while PD has a ratio of 62/38, indicating increased inhibition in the dlPFC. Microglia displayed the most pronounced differences in gene expression profiles between the two conditions. In PD, microglia display a pro-inflammatory phenotype, while in MDD, they regulate synaptic transmission through oligodendrocyte-microglia crosstalk. Analysis of bulk mRNA blood samples revealed that the COL5A, MID1, ZNF148, and CD22 genes were highly expressed in PD, whereas the DENR and RNU1G2 genes were highly expressed in MDD. CD22 is involved in B-cell activation and the negative regulation of B-cell receptor signaling. Additionally, CD86, which provides co-stimulatory signals for T-cell activation and survival, was found to be a commonly differentially expressed gene in both conditions. Pathway analysis revealed several immune-related pathways common in both conditions, including the complement and coagulation cascade, and B-cell receptor signaling. Discussion This study demonstrates that bulk peripheral immune cells play a role in both conditions, but neuroinflammation in the dlPFC specifically manifests in PD as evidenced by the analysis of single nucleus dlPFC datasets. Integrating these two omics levels offers a better understanding of the shared and distinct molecular pathophysiology of PD and MDD in both the periphery and the brain. These findings could lead to potential diagnostic biomarkers, improving accuracy and guiding pharmacological treatments.
Collapse
Affiliation(s)
- Christiana C. Christodoulou
- Neuroepidemiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- The Cyprus Institute of Neurology and Genetics Is a Full Member of the European Reference Network-Rare Neurological Diseases (ERN-RND), Tübingen, Germany
| | - Anna Onisiforou
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Panos Zanos
- Translational Neuropharmacology Laboratory, Department of Psychology, University of Cyprus, Nicosia, Cyprus
| | - Eleni Zamba Papanicolaou
- Neuroepidemiology Department, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
- The Cyprus Institute of Neurology and Genetics Is a Full Member of the European Reference Network-Rare Neurological Diseases (ERN-RND), Tübingen, Germany
| |
Collapse
|
30
|
Litwiniuk A, Juszczak GR, Stankiewicz AM, Urbańska K. The role of glial autophagy in Alzheimer's disease. Mol Psychiatry 2023; 28:4528-4539. [PMID: 37679471 DOI: 10.1038/s41380-023-02242-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/21/2023] [Accepted: 08/25/2023] [Indexed: 09/09/2023]
Abstract
Although Alzheimer's disease is the most pervasive neurodegenerative disorder, the mechanism underlying its development is still not precisely understood. Available data indicate that pathophysiology of this disease may involve impaired autophagy in glial cells. The dysfunction is manifested as reduced ability of astrocytes and microglia to clear abnormal protein aggregates. Consequently, excessive accumulation of amyloid beta plaques and neurofibrillary tangles activates microglia and astrocytes leading to decreased number of mature myelinated oligodendrocytes and death of neurons. These pathologic effects of autophagy dysfunction can be rescued by pharmacological activation of autophagy. Therefore, a deeper understanding of the molecular mechanisms involved in autophagy dysfunction in glial cells in Alzheimer's disease may lead to the development of new therapeutic strategies. However, such strategies need to take into consideration differences in regulation of autophagy in different types of neuroglia.
Collapse
Affiliation(s)
- Anna Litwiniuk
- Department of Neuroendocrinology, Centre of Postgraduate Medical Education, Warsaw, Mazovia, Poland
| | - Grzegorz Roman Juszczak
- Department of Animal Behavior and Welfare, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, Mazovia, Poland
| | - Adrian Mateusz Stankiewicz
- Department of Molecular Biology, Institute of Genetics and Animal Biotechnology, Polish Academy of Sciences, Jastrzębiec, Mazovia, Poland.
| | - Kaja Urbańska
- Department of Morphological Sciences, Faculty of Veterinary Medicine, Warsaw University of Life Sciences, Warsaw, Mazovia, Poland.
| |
Collapse
|
31
|
Chen H, Fan L, Guo Q, Wong MY, Yu F, Foxe N, Wang W, Nessim A, Carling G, Liu B, Lopez-Lee C, Huang Y, Amin S, Patel T, Mok SA, Song WM, Zhang B, Ma Q, Fu H, Gan L, Luo W. DAP12 deficiency alters microglia-oligodendrocyte communication and enhances resilience against tau toxicity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.26.563970. [PMID: 37961594 PMCID: PMC10634844 DOI: 10.1101/2023.10.26.563970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pathogenic tau accumulation fuels neurodegeneration in Alzheimer's disease (AD). Enhancing aging brain's resilience to tau pathology would lead to novel therapeutic strategies. DAP12 (DNAX-activation protein 12) is critically involved in microglial immune responses. Previous studies have showed that mice lacking DAP12 in tauopathy mice exhibit higher tau pathology but are protected from tau-induced cognitive deficits. However, the exact mechanism remains elusive. Our current study uncovers a novel resilience mechanism via microglial interaction with oligodendrocytes. Despite higher tau inclusions, Dap12 deletion curbs tau-induced brain inflammation and ameliorates myelin and synapse loss. Specifically, removal of Dap12 abolished tau-induced disease-associated clusters in microglia (MG) and intermediate oligodendrocytes (iOli), which are spatially correlated with tau pathology in AD brains. Our study highlights the critical role of interactions between microglia and oligodendrocytes in tau toxicity and DAP12 signaling as a promising target for enhancing resilience in AD.
Collapse
Affiliation(s)
- Hao Chen
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Qi Guo
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nessa Foxe
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Aviram Nessim
- The State University of New York at Stony Brook, Long Island, New York, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Tark Patel
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - Sue-Ann Mok
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Millburn High School, New Jersey, NJ, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
32
|
Chen H, Fan L, Guo Q, Wong MY, Yu F, Foxe N, Wang W, Nessim A, Carling G, Liu B, Lopez-Lee C, Huang Y, Amin S, Mok SA, Song WM, Zhang B, Ma Q, Fu H, Gan L, Luo W. DAP12 deficiency alters microglia-oligodendrocyte communication and enhances resilience against tau toxicity. RESEARCH SQUARE 2023:rs.3.rs-3454358. [PMID: 37961627 PMCID: PMC10635319 DOI: 10.21203/rs.3.rs-3454358/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Pathogenic tau accumulation fuels neurodegeneration in Alzheimer's disease (AD). Enhancing aging brain's resilience to tau pathology would lead to novel therapeutic strategies. DAP12 (DNAX-activation protein 12) is critically involved in microglial immune responses. Previous studies have showed that mice lacking DAP12 in tauopathy mice exhibit higher tau pathology but are protected from tau-induced cognitive deficits. However, the exact mechanism remains elusive. Our current study uncovers a novel resilience mechanism via microglial interaction with oligodendrocytes. Despite higher tau inclusions, Dap12 deletion curbs tau-induced brain inflammation and ameliorates myelin and synapse loss. Specifically, removal of Dap12 abolished tau-induced disease-associated clusters in microglia (MG) and intermediate oligodendrocytes (iOli), which are spatially correlated with tau pathology in AD brains. Our study highlights the critical role of interactions between microglia and oligodendrocytes in tau toxicity and DAP12 signaling as a promising target for enhancing resilience in AD.
Collapse
Affiliation(s)
- Hao Chen
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Li Fan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Qi Guo
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Man Ying Wong
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Fangmin Yu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Nessa Foxe
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Aviram Nessim
- The State University of New York at Stony Brook, Long Island, New York, USA
| | - Gillian Carling
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Bangyan Liu
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Chloe Lopez-Lee
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Yige Huang
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Program of Neuroscience, Weill Graduate School of Medical Sciences of Cornell University, New York, NY, USA
| | - Sadaf Amin
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Sue-Ann Mok
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB Canada
| | - Won-min Song
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Bin Zhang
- Department of Genetics and Genomic Sciences, Mount Sinai Center for Transformative Disease Modeling, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Qin Ma
- Department of Biomedical Informatics, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Hongjun Fu
- Department of Neuroscience, College of Medicine, Ohio State University, Columbus, OH 43210 USA
| | - Li Gan
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
- Millburn High School, New Jersey, NJ, USA
| | - Wenjie Luo
- Helen and Robert Appel Alzheimer Disease Research Institute, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
33
|
Kaplanis SI, Kaffe D, Ktena N, Lygeraki A, Kolliniati O, Savvaki M, Karagogeos D. Nicotinamide enhances myelin production after demyelination through reduction of astrogliosis and microgliosis. Front Cell Neurosci 2023; 17:1201317. [PMID: 37663127 PMCID: PMC10469866 DOI: 10.3389/fncel.2023.1201317] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Caloric restriction is the chronic reduction of total caloric intake without malnutrition and has attracted a lot of attention as, among multiple other effects, it attenuates demyelination and stimulates remyelination. In this study we have evaluated the effect of nicotinamide (NAM), a well-known caloric restriction mimetic, on myelin production upon demyelinating conditions. NAM is the derivative of nicotinic acid (vitamin B3) and a precursor of nicotinamide adenine dinucleotide (NAD+), a ubiquitous metabolic cofactor. Here, we use cortical slices ex vivo subjected to demyelination or cultured upon normal conditions, a lysolecithin (LPC)-induced focal demyelination mouse model as well as primary glial cultures. Our data show that NAM enhances both myelination and remyelination ex vivo, while it also induces myelin production after LPC-induced focal demyelination ex vivo and in vivo. The increased myelin production is accompanied by reduction in both astrogliosis and microgliosis in vivo. There is no direct effect of NAM on the oligodendrocyte lineage, as no differences are observed in oligodendrocyte precursor cell proliferation or differentiation or in the number of mature oligodendrocytes. On the other hand, NAM affects both microglia and astrocytes as it decreases the population of M1-activated microglia, while reducing the pro-inflammatory phenotype of astrocytes as assayed by the reduction of TNF-α. Overall, we show that the increased myelin production that follows NAM treatment in vivo is accompanied by a decrease in both astrocyte and microglia accumulation at the lesion site. Our data indicate that NAM influences astrocytes and microglia directly, in favor of the remyelination process by promoting a less inflammatory environment.
Collapse
Affiliation(s)
- Stefanos Ioannis Kaplanis
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Despoina Kaffe
- Department of Biology, University of Crete, Heraklion, Greece
| | - Niki Ktena
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | | | - Ourania Kolliniati
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
- Laboratory of Clinical Chemistry, Medical School, University of Crete, Heraklion, Greece
- Department of Pediatrics, Medical School, University of Crete, Heraklion, Greece
| | - Maria Savvaki
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| | - Domna Karagogeos
- Department of Basic Science, School of Medicine, University of Crete, Heraklion, Greece
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology Hellas, Heraklion, Greece
| |
Collapse
|
34
|
Nguyen JN, Chauhan A. Bystanders or not? Microglia and lymphocytes in aging and stroke. Neural Regen Res 2023; 18:1397-1403. [PMID: 36571333 PMCID: PMC10075112 DOI: 10.4103/1673-5374.360345] [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: 09/17/2022] [Revised: 10/18/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022] Open
Abstract
As the average age of the world population increases, more people will face debilitating aging-associated conditions, including dementia and stroke. Not only does the incidence of these conditions increase with age, but the recovery afterward is often worse in older patients. Researchers and health professionals must unveil and understand the factors behind age-associated diseases to develop a therapy for older patients. Aging causes profound changes in the immune system including the activation of microglia in the brain. Activated microglia promote T lymphocyte transmigration leading to an increase in neuroinflammation, white matter damage, and cognitive impairment in both older humans and rodents. The presence of T and B lymphocytes is observed in the aged brain and correlates with worse stroke outcomes. Preclinical strategies in stroke target either microglia or the lymphocytes or the communications between them to promote functional recovery in aged subjects. In this review, we examine the role of the microglia and T and B lymphocytes in aging and how they contribute to cognitive impairment. Additionally, we provide an important update on the contribution of these cells and their interactions in preclinical aged stroke.
Collapse
Affiliation(s)
- Justin N. Nguyen
- University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| |
Collapse
|
35
|
Zhang X, Chen F, Sun M, Wu N, Liu B, Yi X, Ge R, Fan X. Microglia in the context of multiple sclerosis. Front Neurol 2023; 14:1157287. [PMID: 37360338 PMCID: PMC10287974 DOI: 10.3389/fneur.2023.1157287] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/10/2023] [Indexed: 06/28/2023] Open
Abstract
Multiple sclerosis (MS) is an inflammatory and neurodegenerative disease that commonly results in nontraumatic disability in young adults. The characteristic pathological hallmark of MS is damage to myelin, oligodendrocytes, and axons. Microglia provide continuous surveillance in the CNS microenvironment and initiate defensive mechanisms to protect CNS tissue. Additionally, microglia participate in neurogenesis, synaptic refinement, and myelin pruning through the expression and release of different signaling factors. Continuous activation of microglia has been implicated in neurodegenerative disorders. We first review the lifetime of microglia, including the origin, differentiation, development, and function of microglia. We then discuss microglia participate in the whole processes of remyelination and demyelination, microglial phenotypes in MS, and the NF-κB/PI3K-AKT signaling pathway in microglia. The damage to regulatory signaling pathways may change the homeostasis of microglia, which would accelerate the progression of MS.
Collapse
Affiliation(s)
- Xue Zhang
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Fang Chen
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Mingyue Sun
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Nan Wu
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Bin Liu
- Institute for Metabolic and Neuropsychiatric Disorders, Binzhou Medical University Hospital, Binzhou, China
| | - Xiangming Yi
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Ruli Ge
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| | - Xueli Fan
- Department of Neurology, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
36
|
Fabbri R, Spennato D, Conte G, Konstantoulaki A, Lazzarini C, Saracino E, Nicchia GP, Frigeri A, Zamboni R, Spray DC, Benfenati V. The emerging science of Glioception: Contribution of glia in sensing, transduction, circuit integration of interoception. Pharmacol Ther 2023; 245:108403. [PMID: 37024060 DOI: 10.1016/j.pharmthera.2023.108403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
Interoception is the process by which the nervous system regulates internal functions to achieve homeostasis. The role of neurons in interoception has received considerable recent attention, but glial cells also contribute. Glial cells can sense and transduce signals including osmotic, chemical, and mechanical status of extracellular milieu. Their ability to dynamically communicate "listening" and "talking" to neurons is necessary to monitor and regulate homeostasis and information integration in the nervous system. This review introduces the concept of "Glioception" and focuses on the process by which glial cells sense, interpret and integrate information about the inner state of the organism. Glial cells are ideally positioned to act as sensors and integrators of diverse interoceptive signals and can trigger regulatory responses via modulation of the activity of neuronal networks, both in physiological and pathological conditions. We believe that understanding and manipulating glioceptive processes and underlying molecular mechanisms provide a key path to develop new therapies for the prevention and alleviation of devastating interoceptive dysfunctions, among which pain is emphasized here with more focused details.
Collapse
Affiliation(s)
- Roberta Fabbri
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Electrical, Electronic, and Information Engineering "Guglielmo Marconi", University of Bologna, viale del Risorgimento 2, 40136 Bologna, Italy.
| | - Diletta Spennato
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Giorgia Conte
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Aikaterini Konstantoulaki
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy; Department of Chemistry "Giacomo Ciamician", University of Bologna, Via Selmi, 2, 40126 Bologna, BO, Italy
| | - Chiara Lazzarini
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Emanuela Saracino
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - Grazia Paola Nicchia
- School of Medicine, Basic Medical Sciences, Neuroscience and Sense Organs, University of Bari "Aldo Moro", Bari, BA, Italy; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Antonio Frigeri
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Bioscience, Biotechnologies and Biopharmaceutics, Centre of Excellence in Comparative Genomics, University of Bari "Aldo Moro", Bari, BA, Italy
| | - Roberto Zamboni
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy
| | - David C Spray
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Valentina Benfenati
- Institute for Organic Synthesis and Photoreactivity (ISOF), National Research Council of Italy (CNR), Via P. Gobetti 101, I-40129 Bologna, Italy.
| |
Collapse
|
37
|
Gong X, Gui Z, Ye X, Li X. Jatrorrhizine ameliorates Schwann cell myelination via inhibiting HDAC3 ability to recruit Atxn2l for regulating the NRG1-ErbB2-PI3K-AKT pathway in diabetic peripheral neuropathy mice. Phytother Res 2023; 37:645-657. [PMID: 36218239 DOI: 10.1002/ptr.7641] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 09/04/2022] [Accepted: 09/14/2022] [Indexed: 11/11/2022]
Abstract
Diabetic peripheral neuropathy (DPN) is a chronic complication associated with nerve dysfunction and uncontrolled hyperglycemia. Unfortunately, due to its complicated etiology, there has been no successful therapy for DPN. Our research recently revealed that jatrorrhizine (JAT), one of the active constituents of Rhizoma Coptidis, remarkably ameliorated DPN. This work highlighted the potential mechanism through which JAT relieves DPN using db/db mice. The results indicated that JAT treatment significantly decreased the threshold for thermal and mechanical stimuli and increased nerve conduction velocity. Histopathological analysis revealed that JAT significantly increased the number of sciatic nerve fibers and axons, myelin thickness, and axonal diameters. Additionally, JAT markedly elevated the expression of myelination-associated proteins (MBP, MPZ, and Pmp22). The screening of histone deacetylases (HDAC) determined that histone deacetylase 3 (HDAC3) is an excellent target for JAT-induced myelination enhancement. Liquid chromatography-mass spectrometry-(MS)/MS and coimmunoprecipitation analyses further confirmed that HDAC3 antagonizes the NRG1-ErbB2-PI3K-AKT signaling axis by interacting with Atxn2l to augment SCs myelination. Thus, JAT ameliorates SCs myelination in DPN mice via inhibiting the recruitment of Atxn2l by HDAC3 to regulate the NRG1-ErbB2-PI3K-AKT pathway.
Collapse
Affiliation(s)
- Xiaobao Gong
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| | - Zhenwei Gui
- School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Xiaoli Ye
- School of Life Sciences, Southwest University, Chongqing, People's Republic of China
| | - Xuegang Li
- Engineering Research Center of Coptis Development and Utilization (Ministry of Education), College of Pharmaceutical Sciences, Southwest University, Chongqing, People's Republic of China
| |
Collapse
|
38
|
Maurya SK, Gupta S, Mishra R. Transcriptional and epigenetic regulation of microglia in maintenance of brain homeostasis and neurodegeneration. Front Mol Neurosci 2023; 15:1072046. [PMID: 36698776 PMCID: PMC9870594 DOI: 10.3389/fnmol.2022.1072046] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 12/15/2022] [Indexed: 01/12/2023] Open
Abstract
The emerging role of microglia in brain homeostasis, neurodegeneration, and neurodevelopmental disorders has attracted considerable interest. In addition, recent developments in microglial functions and associated pathways have shed new light on their fundamental role in the immunological surveillance of the brain. Understanding the interconnections between microglia, neurons, and non-neuronal cells have opened up additional avenues for research in this evolving field. Furthermore, the study of microglia at the transcriptional and epigenetic levels has enhanced our knowledge of these native brain immune cells. Moreover, exploring various facets of microglia biology will facilitate the early detection, treatment, and management of neurological disorders. Consequently, the present review aimed to provide comprehensive insight on microglia biology and its influence on brain development, homeostasis, management of disease, and highlights microglia as potential therapeutic targets in neurodegenerative and neurodevelopmental diseases.
Collapse
Affiliation(s)
- Shashank Kumar Maurya
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, University of Delhi, New Delhi, India,*Correspondence: Shashank Kumar Maurya, ;
| | - Suchi Gupta
- Tech Cell Innovations Private Limited, Centre for Medical Innovation and Entrepreneurship (CMIE), All India Institute of Medical Sciences, New Delhi, India
| | - Rajnikant Mishra
- Biochemistry and Molecular Biology Laboratory, Department of Zoology, Banaras Hindu University, Varanasi, India
| |
Collapse
|
39
|
Huang T, Wu J, Mu J, Gao J. Advanced Therapies for Traumatic Central Nervous System Injury: Delivery Strategy Reinforced Efficient Microglial Manipulation. Mol Pharm 2023; 20:41-56. [PMID: 36469398 DOI: 10.1021/acs.molpharmaceut.2c00605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Traumatic central nervous system (CNS) injuries, including spinal cord injury and traumatic brain injury, are challenging enemies of human health. Microglia, the main component of the innate immune system in CNS, can be activated postinjury and are key participants in the pathological procedure and development of CNS trauma. Activated microglia can be typically classified into pro-inflammatory (M1) and anti-inflammatory (M2) phenotypes. Reducing M1 polarization while promoting M2 polarization is thought to be promising for CNS injury treatment. However, obstacles such as the low permeability of the blood-brain barrier and short retention time in circulation limit the therapeutic outcomes of administrated drugs, and rational delivery strategies are necessary for efficient microglial regulation. To this end, proper administration methods and delivery systems like nano/microcarriers and scaffolds are investigated to augment the therapeutic effects of drugs, while some of these delivery systems have self-efficacies in microglial manipulation. Besides, systems based on cell and cell-derived exosomes also show impressive effects, and some underlying targeting mechanisms of these delivery systems have been discovered. In this review, we introduce the roles of microglia play in traumatic CNS injuries, discuss the potential targets for the polarization regulation of microglial phenotype, and summarize recent studies and clinical trials about delivery strategies on enhancing the effect of microglial regulation and therapeutic outcome, as well as targeting mechanisms post CNS trauma.
Collapse
Affiliation(s)
- Tianchen Huang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jiahe Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Department of Clinical Pharmacology, Key Laboratory of Clinical Cancer, Pharmacology and Toxicology Research of Zhejiang Province, Affiliated, Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, China
| | - Jiafu Mu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Hangzhou Institute of Innovative Medicine, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.,Jinhua Institute of Zhejiang University, Jinhua 321002, China
| |
Collapse
|
40
|
Murray CJ, Vecchiarelli HA, Tremblay MÈ. Enhancing axonal myelination in seniors: A review exploring the potential impact cannabis has on myelination in the aged brain. Front Aging Neurosci 2023; 15:1119552. [PMID: 37032821 PMCID: PMC10073480 DOI: 10.3389/fnagi.2023.1119552] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/22/2023] [Indexed: 04/11/2023] Open
Abstract
Consumption of cannabis is on the rise as public opinion trends toward acceptance and its consequent legalization. Specifically, the senior population is one of the demographics increasing their use of cannabis the fastest, but research aimed at understanding cannabis' impact on the aged brain is still scarce. Aging is characterized by many brain changes that slowly alter cognitive ability. One process that is greatly impacted during aging is axonal myelination. The slow degradation and loss of myelin (i.e., demyelination) in the brain with age has been shown to associate with cognitive decline and, furthermore, is a common characteristic of numerous neurological diseases experienced in aging. It is currently not known what causes this age-dependent degradation, but it is likely due to numerous confounding factors (i.e., heightened inflammation, reduced blood flow, cellular senescence) that impact the many cells responsible for maintaining overall homeostasis and myelin integrity. Importantly, animal studies using non-human primates and rodents have also revealed demyelination with age, providing a reliable model for researchers to try and understand the cellular mechanisms at play. In rodents, cannabis was recently shown to modulate the myelination process. Furthermore, studies looking at the direct modulatory impact cannabis has on microglia, astrocytes and oligodendrocyte lineage cells hint at potential mechanisms to prevent some of the more damaging activities performed by these cells that contribute to demyelination in aging. However, research focusing on how cannabis impacts myelination in the aged brain is lacking. Therefore, this review will explore the evidence thus far accumulated to show how cannabis impacts myelination and will extrapolate what this knowledge may mean for the aged brain.
Collapse
Affiliation(s)
- Colin J. Murray
- Neuroscience Graduate Program, University of Victoria, Victoria, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- *Correspondence: Colin J. Murray,
| | | | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Départment de Médicine Moléculaire, Université Laval, Québec City, QC, Canada
- Axe Neurosciences, Center de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Neurology and Neurosurgery Department, McGill University, Montréal, QC, Canada
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Institute for Aging and Lifelong Health, University of Victoria, Victoria, BC, Canada
- Marie-Ève Tremblay,
| |
Collapse
|
41
|
Wang X, Eguchi A, Yang Y, Chang L, Wan X, Shan J, Qu Y, Ma L, Mori C, Yang J, Hashimoto K. Key role of the gut-microbiota-brain axis via the subdiaphragmatic vagus nerve in demyelination of the cuprizone-treated mouse brain. Neurobiol Dis 2023; 176:105951. [PMID: 36493975 DOI: 10.1016/j.nbd.2022.105951] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/22/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Multiple sclerosis (MS) is the most common demyelinating disease that attacks the central nervous system. Dietary intake of cuprizone (CPZ) produces demyelination resembling that of patients with MS. Given the role of the vagus nerve in gut-microbiota-brain axis in development of MS, we performed this study to investigate whether subdiaphragmatic vagotomy (SDV) affects demyelination in CPZ-treated mice. SDV significantly ameliorated demyelination and microglial activation in the brain compared with sham-operated CPZ-treated mice. Furthermore, 16S ribosomal RNA analysis revealed that SDV significantly improved the abnormal gut microbiota composition of CPZ-treated mice. An untargeted metabolomic analysis demonstrated that SDV significantly improved abnormal blood levels of metabolites in CPZ-treated mice compared with sham-operated CPZ-treated mice. Notably, there were correlations between demyelination or microglial activation in the brain and the relative abundance of several microbiome populations, suggesting a link between gut microbiota and the brain. There were also correlations between demyelination or microglial activation in the brain and blood levels of metabolites. Together, these data suggest that CPZ produces demyelination in the brain through the gut-microbiota-brain axis via the subdiaphragmatic vagus nerve.
Collapse
Affiliation(s)
- Xingming Wang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan; Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Akifumi Eguchi
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba 263-8522, Japan
| | - Yong Yang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Lijia Chang
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Xiayun Wan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Jiajing Shan
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Youge Qu
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Li Ma
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan
| | - Chisato Mori
- Department of Sustainable Health Science, Chiba University Center for Preventive Medical Sciences, Chiba 263-8522, Japan; Department of Bioenvironmental Medicine, Graduate School of Medicine, Chiba University, Chiba 260-8670, Japan
| | - Jianjun Yang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan, China
| | - Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Chiba 260-8670, Japan.
| |
Collapse
|
42
|
Picard K, Corsi G, Decoeur F, Di Castro MA, Bordeleau M, Persillet M, Layé S, Limatola C, Tremblay MÈ, Nadjar A. Microglial homeostasis disruption modulates non-rapid eye movement sleep duration and neuronal activity in adult female mice. Brain Behav Immun 2023; 107:153-164. [PMID: 36202169 DOI: 10.1016/j.bbi.2022.09.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 09/12/2022] [Accepted: 09/30/2022] [Indexed: 11/07/2022] Open
Abstract
Sleep is a natural physiological state, tightly regulated through several neuroanatomical and neurochemical systems, which is essential to maintain physical and mental health. Recent studies revealed that the functions of microglia, the resident immune cells of the brain, differ along the sleep-wake cycle. Inflammatory cytokines, such as interleukin-1β and tumor necrosis factor-α, mainly produced by microglia in the brain, are also well-known to promote sleep. However, the contributing role of microglia on sleep regulation remains largely elusive, even more so in females. Given the higher prevalence of various sleep disorders in women, we aimed to determine the role of microglia in regulating the sleep-wake cycle specifically in female mice. Microglia were depleted in adult female mice with inhibitors of the colony-stimulating factor 1 receptor (CSF1R) (PLX3397 or PLX5622), which is required for microglial population maintenance. This led to a 65-73% reduction of the microglial population, as confirmed by immunofluorescence staining against IBA1 (marker of microglia/macrophages) and TMEM119 (microglia-specific marker) in the reticular nucleus of the thalamus and primary motor cortex. The spontaneous sleep-wake cycle was evaluated at steady-state, during microglial homeostasis disruption and after complete microglial repopulation, upon cessation of treatment with the inhibitors of CSF1R, using electroencephalography (EEG) and electromyography (EMG). We found that microglia-depleted female mice spent more time in non-rapid eye movement (NREM) sleep and had an increased number of NREM sleep episodes, which was partially restored after microglial total repopulation. To determine whether microglia could regulate sleep locally by modulating synaptic transmission, we used patch clamp to record spontaneous activity of pyramidal neurons in the primary motor cortex, which showed an increase of excitatory synaptic transmission during the dark phase. These changes in neuronal activity were modulated by microglial depletion in a phase-dependent manner. Altogether, our results indicate that microglia are involved in the sleep regulation of female mice, further strengthening their potential implication in the development and/or progression of sleep disorders. Furthermore, our findings indicate that microglial repopulation can contribute to normalizing sleep alterations caused by their partial depletion.
Collapse
Affiliation(s)
- Katherine Picard
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Giorgio Corsi
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy
| | - Fanny Decoeur
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | | | - Maude Bordeleau
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Marine Persillet
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Sophie Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France
| | - Cristina Limatola
- Department of Physiology and Pharmacology, Sapienza University of Rome, Rome, Italy; Department of Neurophysiology, Neuropharmacology, Inflammaging, IRCCS Neuromed, Pozzilli, Italy
| | - Marie-Ève Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Département de médecine moléculaire, Université Laval, Québec, QC, Canada; Division of Medical Sciences, University of Victoria, Victoria, BC, Canada; Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada; Department of Neurology and Neurosurgery, McGill University, Montréal, QC, Canada; Centre for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada.
| | - Agnès Nadjar
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, F-33000 Bordeaux, France; INSERM, Neurocentre Magendie, Physiopathologie de la Plasticité Neuronale, U1215, F-33000 Bordeaux, France; Institut Universitaire de France (IUF), France.
| |
Collapse
|
43
|
Deciphering the Genetic Crosstalk between Microglia and Oligodendrocyte Precursor Cells during Demyelination and Remyelination Using Transcriptomic Data. Int J Mol Sci 2022; 23:ijms232314868. [PMID: 36499195 PMCID: PMC9738937 DOI: 10.3390/ijms232314868] [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: 09/22/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
Demyelinating disorders show impaired remyelination due to failure in the differentiation of oligodendrocyte progenitor cells (OPCs) into mature myelin-forming oligodendrocytes, a process driven by microglia-OPC crosstalk. Through conducting a transcriptomic analysis of microarray studies on the demyelination-remyelination cuprizone model and using human samples of multiple sclerosis (MS), we identified molecules involved in this crosstalk. Differentially expressed genes (DEGs) of specific regions/cell types were detected in GEO transcriptomic raw data after cuprizone treatment and in MS samples, followed by functional analysis with GO terms and WikiPathways. Additionally, microglia-OPC crosstalk between microglia ligands, OPC receptors and target genes was examined with the NicheNet model. We identified 108 and 166 DEGs in the demyelinated corpus callosum (CC) at 2 and 4 weeks of cuprizone treatment; 427 and 355 DEGs in the remyelinated (4 weeks of cuprizone treatment + 14 days of normal diet) compared to 2- and 4-week demyelinated CC; 252 DEGs in MS samples and 2730 and 12 DEGs in OPC and microglia of 4-week demyelinated CC. At this time point, we found 95 common DEGs in the CC and OPCs, and one common DEG in microglia and OPCs, mostly associated with myelin and lipid metabolism. Crosstalk analysis identified 47 microglia ligands, 43 OPC receptors and 115 OPC target genes, all differentially expressed in cuprizone-treated samples and associated with myelination. Our differential expression pipeline identified demyelination/remyelination transcriptomic biomarkers in studies using diverse platforms and cell types/tissues. Cellular crosstalk analysis yielded novel markers of microglia ligands, OPC receptors and target genes.
Collapse
|
44
|
Carrier M, Dolhan K, Bobotis BC, Desjardins M, Tremblay MÈ. The implication of a diversity of non-neuronal cells in disorders affecting brain networks. Front Cell Neurosci 2022; 16:1015556. [PMID: 36439206 PMCID: PMC9693782 DOI: 10.3389/fncel.2022.1015556] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/07/2022] [Indexed: 11/13/2022] Open
Abstract
In the central nervous system (CNS) neurons are classically considered the functional unit of the brain. Analysis of the physical connections and co-activation of neurons, referred to as structural and functional connectivity, respectively, is a metric used to understand their interplay at a higher level. A myriad of glial cell types throughout the brain composed of microglia, astrocytes and oligodendrocytes are key players in the maintenance and regulation of neuronal network dynamics. Microglia are the central immune cells of the CNS, able to affect neuronal populations in number and connectivity, allowing for maturation and plasticity of the CNS. Microglia and astrocytes are part of the neurovascular unit, and together they are essential to protect and supply nutrients to the CNS. Oligodendrocytes are known for their canonical role in axonal myelination, but also contribute, with microglia and astrocytes, to CNS energy metabolism. Glial cells can achieve this variety of roles because of their heterogeneous populations comprised of different states. The neuroglial relationship can be compromised in various manners in case of pathologies affecting development and plasticity of the CNS, but also consciousness and mood. This review covers structural and functional connectivity alterations in schizophrenia, major depressive disorder, and disorder of consciousness, as well as their correlation with vascular connectivity. These networks are further explored at the cellular scale by integrating the role of glial cell diversity across the CNS to explain how these networks are affected in pathology.
Collapse
Affiliation(s)
- Micaël Carrier
- Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Kira Dolhan
- Department of Psychology, University of Victoria, Victoria, BC, Canada
- Department of Biology, University of Victoria, Victoria, BC, Canada
| | | | - Michèle Desjardins
- Department of Physics, Physical Engineering and Optics, Université Laval, Québec City, QC, Canada
- Oncology Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
| | - Marie-Ève Tremblay
- Neurosciences Axis, Centre de Recherche du CHU de Québec, Université Laval, Québec City, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Department of Molecular Medicine, Université Laval, Québec City, QC, Canada
- Department of Biochemistry and Molecular Biology, The University of British Columbia, Vancouver, BC, Canada
- *Correspondence: Marie-Ève Tremblay,
| |
Collapse
|
45
|
Myatich A, Haque A, Sole C, Banik NL. Clemastine in remyelination and protection of neurons and skeletal muscle after spinal cord injury. Neural Regen Res 2022; 18:940-946. [PMID: 36254972 PMCID: PMC9827778 DOI: 10.4103/1673-5374.355749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Spinal cord injuries affect nearly five to ten individuals per million every year. Spinal cord injury causes damage to the nerves, muscles, and the tissue surrounding the spinal cord. Depending on the severity, spinal injuries are linked to degeneration of axons and myelin, resulting in neuronal impairment and skeletal muscle weakness and atrophy. The protection of neurons and promotion of myelin regeneration during spinal cord injury is important for recovery of function following spinal cord injury. Current treatments have little to no effect on spinal cord injury and neurogenic muscle loss. Clemastine, an Food and Drug Administration-approved antihistamine drug, reduces inflammation, protects cells, promotes remyelination, and preserves myelin integrity. Recent clinical evidence suggests that clemastine can decrease the loss of axons after spinal cord injury, stimulating the differentiation of oligodendrocyte progenitor cells into mature oligodendrocytes that are capable of myelination. While clemastine can aid not only in the remyelination and preservation of myelin sheath integrity, it also protects neurons. However, its role in neurogenic muscle loss remains unclear. This review discusses the pathophysiology of spinal cord injury, and the role of clemastine in the protection of neurons, myelin, and axons as well as attenuation of skeletal muscle loss following spinal cord injury.
Collapse
Affiliation(s)
- Ali Myatich
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA,Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA,Correspondence to: Azizul Haque, ; Naren L. Banik, .
| | - Christopher Sole
- Department of Health and Human Performance, The Citadel, Charleston, SC, USA
| | - Naren L. Banik
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, USA,Department of Neurosurgery, Medical University of South Carolina, Charleston, SC, USA,Ralph H. Johnson Veterans Administration Medical Center, Charleston, SC, USA,Correspondence to: Azizul Haque, ; Naren L. Banik, .
| |
Collapse
|
46
|
Hu J, Baydyuk M, Huang JK. Impact of amino acids on microglial activation and CNS remyelination. Curr Opin Pharmacol 2022; 66:102287. [PMID: 36067684 DOI: 10.1016/j.coph.2022.102287] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 07/20/2022] [Accepted: 08/01/2022] [Indexed: 11/27/2022]
Abstract
Amino acids and their derivatives function as building blocks as well as signaling molecules to modulate various cellular processes in living organisms. In mice, amino acids accumulate in demyelinated lesions and return to basal levels during remyelination. Studies have found that amino acids and their metabolites modulate immune activity in the central nervous system (CNS) and influence oligodendrocyte differentiation and remyelination efficiency. In this review, we discuss current studies on amino acid metabolism in the context of CNS remyelination. By understanding the mechanisms of amino acid signaling and metabolism in demyelinated lesions, we may deepen our understanding of compartmentalized CNS inflammation in demyelinating disease like multiple sclerosis (MS) and provide evidence to develop novel pharmacological therapies targeting amino acid metabolism to prevent disease worsening.
Collapse
Affiliation(s)
- Jingwen Hu
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA
| | - Maryna Baydyuk
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA; Center for Cell Reprogramming, Georgetown University Medical Center, 37th and O St., NW, Washington, DC, 20057, USA
| | - Jeffrey K Huang
- Department of Biology, Georgetown University, 37th and O St., NW, Washington, DC, 20057, USA; Center for Cell Reprogramming, Georgetown University Medical Center, 37th and O St., NW, Washington, DC, 20057, USA.
| |
Collapse
|
47
|
Tykhonenko T, Guzyk M, Tykhomyrov A, Korsa V, Yanitska L, Kuchmerovska T. Modulatory effects of vitamin B3 and its derivative on the levels of apoptotic and vascular regulators and cytoskeletal proteins in diabetic rat brain as signs of neuroprotection. Biochim Biophys Acta Gen Subj 2022; 1866:130207. [PMID: 35882257 DOI: 10.1016/j.bbagen.2022.130207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Revised: 07/08/2022] [Accepted: 07/18/2022] [Indexed: 10/31/2022]
Abstract
BACKGROUND Beneficial effects of nicotinamide (NAm) and its derivates have been earlier shown in animal models of diabetes mellitus (DM), but the mechanisms of their neuroprotective activities are still largely unknown. The aim of the present study was to investigate if NAm and conjugate of nicotinic acid with gamma-aminobutyric acid (N-GABA) are able to modulate expression levels of apoptosis regulators, angiogenesis-related molecules, and specific cytoskeletal proteins in diabetic rat brain. METHODS After six weeks of streptozotocin induced type 1 DM, rats were daily administered either by NAm (100 mg/kg) or N-GABA (55 mg/kg) intraperitoneally for two weeks. Protein levels were assessed by western blot and immunohistochemistry. RESULTS Both NAm and N-GABA down-regulated NF-κB and Bax levels in diabetic rat brain, suggesting their anti-apoptotic activities. Tested compounds normalized VEGF and nNOS contents improving pro-angiogenic signaling reduced by hyperglycemia. Western blot showed marked up-regulation of astroglial marker GFAP and lowering neurofilament protein levels in DM group, confirmed immunohistochemically, indicating the development of reactive astrogliosis as a major response to diabetes-induced neurodegeneration. NAm had no effects on GFAP and Nf-L levels in the diabetic brain, while N-GABA increased their expression. Inversely, NAm and N-GABA dramatically reduced enhanced levels of GFAP and Nf-L found in the blood serum of diabetic rats, providing for the first time strong evidence for preserving blood-brain barrier integrity by studied compounds. CONCLUSION Thus, NAm and N-GABA may exert neuroprotective effects by decreasing pro-apoptotic regulators levels and improving expression of angiogenic and cytoskeletal proteins impaired by hyperglycemia in rat brain.
Collapse
Affiliation(s)
- Tetiana Tykhonenko
- Department of Vitamin and Coenzyme Biochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Ukraine
| | - Mykhailo Guzyk
- Department of Vitamin and Coenzyme Biochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Ukraine
| | - Artem Tykhomyrov
- Department of Enzyme Chemistry and Biochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Ukraine
| | - Victoria Korsa
- Department of Enzyme Chemistry and Biochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Ukraine
| | - Lesya Yanitska
- Department of Medical Biochemistry and Molecular Biology, Bogomolets National Medical University, Kyiv, Ukraine
| | - Tamara Kuchmerovska
- Department of Vitamin and Coenzyme Biochemistry, Palladin Institute of Biochemistry, National Academy of Sciences of Ukraine, Ukraine.
| |
Collapse
|
48
|
Kawabe Y, Tanaka T, Isonishi A, Nakahara K, Tatsumi K, Wanaka A. Characterization of Glial Populations in the Aging and Remyelinating Mouse Corpus Callosum. Neurochem Res 2022; 47:2826-2838. [PMID: 35859078 DOI: 10.1007/s11064-022-03676-z] [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: 03/30/2022] [Revised: 06/15/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
Cells in the white matter of the adult brain have a characteristic distribution pattern in which several cells are contiguously connected to each other, making a linear array (LA) resembling pearls-on-a-string parallel to the axon axis. We have been interested in how this pattern of cell distribution changes during aging and remyelination after demyelination. In the present study, with a multiplex staining method, semi-quantitative analysis of the localization of oligodendrocyte lineage cells (oligodendrocyte progenitors, premyelinating oligodendrocytes, and mature oligodendrocytes), astrocytes, and microglia in 8-week-old (young adult) and 32-week-old (aged) corpus callosum showed that young adult cells still include immature oligodendrocytes and that LAs contain a higher proportion of microglia than isolated cells. In aged mice, premyelinating oligodendrocytes were decreased, but microglia continued to be present in the LAs. These results suggest that the presence of microglia is important for the characteristic cell localization pattern of LAs. In a cuprizone-induced demyelination model, we observed re-formation of LAs after completion of cuprizone treatment, concurrent with remyelination. These re-formed LAs again contained more microglia than the isolated cells. This finding supports the hypothesis that microglia contribute to the formation and maintenance of LAs. In addition, regardless of the distribution of cells (LAs or isolated cells), astrocytes were found to be more abundant than in the normal corpus callosum at 24 weeks after cuprizone treatment when remyelination is completed. This suggests that astrocytes are involved in maintaining the functions of remyelinated white matter.
Collapse
Affiliation(s)
- Yoshie Kawabe
- Department of Anatomy and Neuroscience, Nara Medical University, Faculty of Medicine, 840 Shijo-cho, Kashihara City, Nara, 634-8521, Japan
| | - Tatsuhide Tanaka
- Department of Anatomy and Neuroscience, Nara Medical University, Faculty of Medicine, 840 Shijo-cho, Kashihara City, Nara, 634-8521, Japan
| | - Ayami Isonishi
- Department of Anatomy and Neuroscience, Nara Medical University, Faculty of Medicine, 840 Shijo-cho, Kashihara City, Nara, 634-8521, Japan
| | - Kazuki Nakahara
- Department of Anatomy and Neuroscience, Nara Medical University, Faculty of Medicine, 840 Shijo-cho, Kashihara City, Nara, 634-8521, Japan
| | - Kouko Tatsumi
- Department of Anatomy and Neuroscience, Nara Medical University, Faculty of Medicine, 840 Shijo-cho, Kashihara City, Nara, 634-8521, Japan.
| | - Akio Wanaka
- Department of Anatomy and Neuroscience, Nara Medical University, Faculty of Medicine, 840 Shijo-cho, Kashihara City, Nara, 634-8521, Japan
| |
Collapse
|
49
|
Targeting microglia–oligodendrocyte crosstalk in neurodegenerative and psychiatric disorders. Drug Discov Today 2022; 27:2562-2573. [DOI: 10.1016/j.drudis.2022.06.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 06/09/2022] [Accepted: 06/29/2022] [Indexed: 02/07/2023]
|
50
|
Renz-Polster H, Tremblay ME, Bienzle D, Fischer JE. The Pathobiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome: The Case for Neuroglial Failure. Front Cell Neurosci 2022; 16:888232. [PMID: 35614970 PMCID: PMC9124899 DOI: 10.3389/fncel.2022.888232] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/13/2022] [Indexed: 12/20/2022] Open
Abstract
Although myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) has a specific and distinctive profile of clinical features, the disease remains an enigma because causal explanation of the pathobiological matrix is lacking. Several potential disease mechanisms have been identified, including immune abnormalities, inflammatory activation, mitochondrial alterations, endothelial and muscular disturbances, cardiovascular anomalies, and dysfunction of the peripheral and central nervous systems. Yet, it remains unclear whether and how these pathways may be related and orchestrated. Here we explore the hypothesis that a common denominator of the pathobiological processes in ME/CFS may be central nervous system dysfunction due to impaired or pathologically reactive neuroglia (astrocytes, microglia and oligodendrocytes). We will test this hypothesis by reviewing, in reference to the current literature, the two most salient and widely accepted features of ME/CFS, and by investigating how these might be linked to dysfunctional neuroglia. From this review we conclude that the multifaceted pathobiology of ME/CFS may be attributable in a unifying manner to neuroglial dysfunction. Because the two key features - post exertional malaise and decreased cerebral blood flow - are also recognized in a subset of patients with post-acute sequelae COVID, we suggest that our findings may also be pertinent to this entity.
Collapse
Affiliation(s)
- Herbert Renz-Polster
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| | - Marie-Eve Tremblay
- Axe Neurosciences, Centre de recherche du CHU de Québec, Université Laval, Quebec, QC, Canada
- Département de Médecine Moléculaire, Université Laval, Quebec, QC, Canada
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
- Center for Advanced Materials and Related Technology (CAMTEC), University of Victoria, Victoria, BC, Canada
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Dorothee Bienzle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Joachim E. Fischer
- Division of General Medicine, Center for Preventive Medicine and Digital Health Baden-Württemberg (CPD-BW), University Medicine Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|