1
|
Mora P, Laisné M, Bourguignon C, Rouault P, Jaspard-Vinassa B, Maître M, Gadeau AP, Renault MA, Horng S, Couffinhal T, Chapouly C. Astrocytic DLL4-NOTCH1 signaling pathway promotes neuroinflammation via the IL-6-STAT3 axis. J Neuroinflammation 2024; 21:258. [PMID: 39390606 PMCID: PMC11468415 DOI: 10.1186/s12974-024-03246-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 09/26/2024] [Indexed: 10/12/2024] Open
Abstract
Under neuroinflammatory conditions, astrocytes acquire a reactive phenotype that drives acute inflammatory injury as well as chronic neurodegeneration. We hypothesized that astrocytic Delta-like 4 (DLL4) may interact with its receptor NOTCH1 on neighboring astrocytes to regulate astrocyte reactivity via downstream juxtacrine signaling pathways. Here we investigated the role of astrocytic DLL4 on neurovascular unit homeostasis under neuroinflammatory conditions. We probed for downstream effectors of the DLL4-NOTCH1 axis and targeted these for therapy in two models of CNS inflammatory disease. We first demonstrated that astrocytic DLL4 is upregulated during neuroinflammation, both in mice and humans, driving astrocyte reactivity and subsequent blood-brain barrier permeability and inflammatory infiltration. We then showed that the DLL4-mediated NOTCH1 signaling in astrocytes directly drives IL-6 levels, induces STAT3 phosphorylation promoting upregulation of astrocyte reactivity markers, pro-permeability factor secretion and consequent blood-brain barrier destabilization. Finally we revealed that blocking DLL4 with antibodies improves experimental autoimmune encephalomyelitis symptoms in mice, identifying a potential novel therapeutic strategy for CNS autoimmune demyelinating disease. As a general conclusion, this study demonstrates that DLL4-NOTCH1 signaling is not only a key pathway in vascular development and angiogenesis, but also in the control of astrocyte reactivity during neuroinflammation.
Collapse
Affiliation(s)
- Pierre Mora
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Margaux Laisné
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Célia Bourguignon
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Paul Rouault
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Béatrice Jaspard-Vinassa
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Marlène Maître
- Univ. Bordeaux, INSERM, Neurocentre Magendie, U1215, Bordeaux, F-33000, France
| | - Alain-Pierre Gadeau
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Marie-Ange Renault
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Sam Horng
- Department of Neurology and Neuroscience, Icahn School of Medicine at Mount Sinai, New York City, NY, USA
| | - Thierry Couffinhal
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France
| | - Candice Chapouly
- Univ. Bordeaux, INSERM, Biology of Cardiovascular Diseases, U1034, 01 avenue de Magellan, Pessac, 33601, France.
| |
Collapse
|
2
|
Inoue Y, Ren Y, Zhang S, Bamkole M, Islam NN, Selvaraj M, Lu W, Caulfield TR, Li Y, Kanekiyo T. A novel histone deacetylase inhibitor W2A-16 improves the barrier integrity in brain vascular endothelial cells. Front Cell Neurosci 2024; 18:1368018. [PMID: 39100897 PMCID: PMC11294206 DOI: 10.3389/fncel.2024.1368018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 07/08/2024] [Indexed: 08/06/2024] Open
Abstract
The maturation of brain microvascular endothelial cells leads to the formation of a tightly sealed monolayer, known as the blood-brain barrier (BBB). The BBB damage is associated with the pathogenesis of age-related neurodegenerative diseases including vascular cognitive impairment and Alzheimer's disease. Growing knowledge in the field of epigenetics can enhance the understanding of molecular profile of the BBB and has great potential for the development of novel therapeutic strategies or targets to repair a disrupted BBB. Histone deacetylases (HDACs) inhibitors are epigenetic regulators that can induce acetylation of histones and induce open chromatin conformation, promoting gene expression by enhancing the binding of DNA with transcription factors. We investigated how HDAC inhibition influences the barrier integrity using immortalized human endothelial cells (HCMEC/D3) and the human induced pluripotent stem cell (iPSC)-derived brain vascular endothelial cells. The endothelial cells were treated with or without a novel compound named W2A-16. W2A-16 not only activates Wnt/β-catenin signaling but also functions as a class I HDAC inhibitor. We demonstrated that the administration with W2A-16 sustained barrier properties of the monolayer of endothelial cells, as evidenced by increased trans-endothelial electrical resistance (TEER). The BBB-related genes and protein expression were also increased compared with non-treated controls. Analysis of transcript profiles through RNA-sequencing in hCMEC/D3 cells indicated that W2A-16 potentially enhances BBB integrity by influencing genes associated with the regulation of the extracellular microenvironment. These findings collectively propose that the HDAC inhibition by W2A-16 plays a facilitating role in the formation of the BBB. Pharmacological approaches to inhibit HDAC may be a potential therapeutic strategy to boost and/or restore BBB integrity.
Collapse
Affiliation(s)
- Yasuteru Inoue
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Yingxue Ren
- Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Shuwen Zhang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN, United States
| | - Michael Bamkole
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Naeyma N. Islam
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | | | - Wenyan Lu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, United States
| | | | - Yonghe Li
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
| | - Takahisa Kanekiyo
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, United States
- Center for Regenerative Biotherapeutics, Mayo Clinic, Jacksonville, FL, United States
| |
Collapse
|
3
|
Duan M, Ru X, Zhou J, Li Y, Guo P, Kang W, Li W, Chen Z, Feng H, Chen Y. Endothelial EGLN3-PKM2 signaling induces the formation of acute astrocytic barrier to alleviate immune cell infiltration after subarachnoid hemorrhage. Fluids Barriers CNS 2024; 21:42. [PMID: 38755642 PMCID: PMC11100217 DOI: 10.1186/s12987-024-00550-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Accepted: 05/09/2024] [Indexed: 05/18/2024] Open
Abstract
BACKGROUND Most subarachnoid hemorrhage (SAH) patients have no obvious hematoma lesions but exhibit blood-brain barrier dysfunction and vasogenic brain edema. However, there is a few days between blood‒brain barrier dysfunction and vasogenic brain edema. The present study sought to investigate whether this phenomenon is caused by endothelial injury induced by the acute astrocytic barrier, also known as the glial limitans. METHODS Bioinformatics analyses of human endothelial cells and astrocytes under hypoxia were performed based on the GEO database. Wild-type, EGLN3 and PKM2 conditional knock-in mice were used to confirm glial limitan formation after SAH. Then, the effect of endothelial EGLN3-PKM2 signaling on temporal and spatial changes in glial limitans was evaluated in both in vivo and in vitro models of SAH. RESULTS The data indicate that in the acute phase after SAH, astrocytes can form a temporary protective barrier, the glia limitans, around blood vessels that helps maintain barrier function and improve neurological prognosis. Molecular docking studies have shown that endothelial cells and astrocytes can promote glial limitans-based protection against early brain injury through EGLN3/PKM2 signaling and further activation of the PKC/ERK/MAPK signaling pathway in astrocytes after SAH. CONCLUSION Improving the ability to maintain glial limitans may be a new therapeutic strategy for improving the prognosis of SAH patients.
Collapse
Affiliation(s)
- Mingxu Duan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xufang Ru
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jiru Zhou
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yuanshu Li
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wenbo Kang
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Wenyan Li
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhi Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), 29 Gaotanyan Street, Shapingba District, Chongqing, 400038, China.
- Chongqing Key Laboratory of Intelligent Diagnosis, Treatment and Rehabilitation of Central Nervous System Injuries, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
- Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|
4
|
Feng L, Li Y, Lin M, Xie D, Luo Y, Zhang Y, He Z, Gong Q, Zhun ZY, Gao J. Trilobatin attenuates cerebral ischaemia/reperfusion-induced blood-brain barrier dysfunction by targeting matrix metalloproteinase 9: The legend of a food additive. Br J Pharmacol 2024; 181:1005-1027. [PMID: 37723895 DOI: 10.1111/bph.16239] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 08/21/2023] [Accepted: 09/05/2023] [Indexed: 09/20/2023] Open
Abstract
BACKGROUND AND PURPOSE Blood-brain barrier (BBB) breakdown is one of the crucial pathological changes of cerebral ischaemia-reperfusion (I/R) injury. Trilobatin (TLB), a naturally occurring food additive, exerts neuroprotective effects against cerebral I/R injury as demonstrated in our previous study. This study was designed to investigate the effect of TLB on BBB disruption after cerebral I/R injury. EXPERIMENTAL APPROACH Rats with focal cerebral ischaemia caused by transient middle cerebral artery occlusion were studied along with brain microvascular endothelial cells and human astrocytes to mimic BBB injury caused by oxygen and glucose deprivation/reoxygenation (OGD/R). KEY RESULTS The results showed that TLB effectively maintained BBB integrity and inhibited neuronal loss following cerebral I/R challenge. Furthermore, TLB increased tight junction proteins including ZO-1, Occludin and Claudin 5, and decreased the levels of apolipoprotein E (APOE) 4, cyclophilin A (CypA) and phosphorylated nuclear factor kappa B (NF-κB), thereby reducing proinflammatory cytokines. TLB also decreased the Bax/Bcl-2 ratio and cleaved-caspase 3 levels along with a reduced number of apoptotic neurons. Molecular docking and transcriptomics predicted MMP9 as a prominent gene evoked by TLB treatment. The protective effects of TLB on cerebral I/R-induced BBB breakdown was largely abolished by overexpression of MMP9, and the beneficial effects of TLB on OGD/R-induced loss of BBB integrity in human brain microvascular endothelial cells and astrocyte co-cultures was markedly reinforced by knockdown of MMP9. CONCLUSIONS AND IMPLICATIONS Our findings reveal a novel property of TLB: preventing BBB disruption following cerebral I/R via targeting MMP9 and inhibiting APOE4/CypA/NF-κB axis.
Collapse
Affiliation(s)
- Linying Feng
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yeli Li
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Mu Lin
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Dianyou Xie
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yunmei Luo
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Yuandong Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Zhixu He
- The Collaborative Innovation Center of Tissue Damage Repair and Regeneration Medicine of Zunyi Medical University, Zunyi, China
| | - Qihai Gong
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| | - Zhu Yi Zhun
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
| | - Jianmei Gao
- School of Pharmacy, Faculty of Medicine, Macau University of Science and Technology, Macau SAR, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
| |
Collapse
|
5
|
He L, Zhang R, Yang M, Lu M. The role of astrocyte in neuroinflammation in traumatic brain injury. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166992. [PMID: 38128844 DOI: 10.1016/j.bbadis.2023.166992] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/30/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
Traumatic brain injury (TBI), a significant contributor to mortality and morbidity worldwide, is a devastating condition characterized by initial mechanical damage followed by subsequent biochemical processes, including neuroinflammation. Astrocytes, the predominant glial cells in the central nervous system, play a vital role in maintaining brain homeostasis and supporting neuronal function. Nevertheless, in response to TBI, astrocytes undergo substantial phenotypic alternations and actively contribute to the neuroinflammatory response. This article explores the multifaceted involvement of astrocytes in neuroinflammation subsequent to TBI, with a particular emphasis on their activation, release of inflammatory mediators, modulation of the blood-brain barrier, and interactions with other immune cells. A comprehensive understanding the dynamic interplay between astrocytes and neuroinflammation in the condition of TBI can provide valuable insights into the development of innovative therapeutic approaches aimed at mitigating secondary damage and fostering neuroregeneration.
Collapse
Affiliation(s)
- Liang He
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China.
| | - Ruqiang Zhang
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Maiqiao Yang
- Department of Anesthesiology, Yan'an Hospital of Kunming City, Kunming 650051, China
| | - Meilin Lu
- Department of Anesthesiology, First Affiliated Hospital of Kunming Medical University, Kunming 650032, China.
| |
Collapse
|
6
|
Pan Y, Li J, Lin P, Wan L, Qu Y, Cao L, Wang L. A review of the mechanisms of abnormal ceramide metabolism in type 2 diabetes mellitus, Alzheimer's disease, and their co-morbidities. Front Pharmacol 2024; 15:1348410. [PMID: 38379904 PMCID: PMC10877008 DOI: 10.3389/fphar.2024.1348410] [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: 12/02/2023] [Accepted: 01/18/2024] [Indexed: 02/22/2024] Open
Abstract
The global prevalence of type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) is rapidly increasing, revealing a strong association between these two diseases. Currently, there are no curative medication available for the comorbidity of T2DM and AD. Ceramides are structural components of cell membrane lipids and act as signal molecules regulating cell homeostasis. Their synthesis and degradation play crucial roles in maintaining metabolic balance in vivo, serving as important mediators in the development of neurodegenerative and metabolic disorders. Abnormal ceramide metabolism disrupts intracellular signaling, induces oxidative stress, activates inflammatory factors, and impacts glucose and lipid homeostasis in metabolism-related tissues like the liver, skeletal muscle, and adipose tissue, driving the occurrence and progression of T2DM. The connection between changes in ceramide levels in the brain, amyloid β accumulation, and tau hyper-phosphorylation is evident. Additionally, ceramide regulates cell survival and apoptosis through related signaling pathways, actively participating in the occurrence and progression of AD. Regulatory enzymes, their metabolites, and signaling pathways impact core pathological molecular mechanisms shared by T2DM and AD, such as insulin resistance and inflammatory response. Consequently, regulating ceramide metabolism may become a potential therapeutic target and intervention for the comorbidity of T2DM and AD. The paper comprehensively summarizes and discusses the role of ceramide and its metabolites in the pathogenesis of T2DM and AD, as well as the latest progress in the treatment of T2DM with AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Lei Wang
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| |
Collapse
|
7
|
Rouault P, Guimbal S, Cornuault L, Bourguignon C, Foussard N, Alzieu P, Choveau F, Benoist D, Chapouly C, Gadeau AP, Couffinhal T, Renault MA. Thrombosis in the Coronary Microvasculature Impairs Cardiac Relaxation and Induces Diastolic Dysfunction. Arterioscler Thromb Vasc Biol 2024; 44:e1-e18. [PMID: 38031839 DOI: 10.1161/atvbaha.123.320040] [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: 08/22/2023] [Accepted: 11/14/2023] [Indexed: 12/01/2023]
Abstract
BACKGROUND Heart failure with preserved ejection fraction is proposed to be caused by endothelial dysfunction in cardiac microvessels. Our goal was to identify molecular and cellular mechanisms underlying the development of cardiac microvessel disease and diastolic dysfunction in the setting of type 2 diabetes. METHODS We used Leprdb/db (leptin receptor-deficient) female mice as a model of type 2 diabetes and heart failure with preserved ejection fraction and identified Hhipl1 (hedgehog interacting protein-like 1), which encodes for a decoy receptor for HH (hedgehog) ligands as a gene upregulated in the cardiac vascular fraction of diseased mice. RESULTS We then used Dhh (desert HH)-deficient mice to investigate the functional consequences of impaired HH signaling in the adult heart. We found that Dhh-deficient mice displayed increased end-diastolic pressure while left ventricular ejection fraction was comparable to that of control mice. This phenotype was associated with a reduced exercise tolerance in the treadmill test, suggesting that Dhh-deficient mice do present heart failure. At molecular and cellular levels, impaired cardiac relaxation in DhhECKO mice was associated with a significantly decreased PLN (phospholamban) phosphorylation on Thr17 (threonine 17) and an alteration of sarcomeric shortening ex vivo. Besides, as expected, Dhh-deficient mice exhibited phenotypic changes in their cardiac microvessels including a prominent prothrombotic phenotype. Importantly, aspirin therapy prevented the occurrence of both diastolic dysfunction and exercise intolerance in these mice. To confirm the critical role of thrombosis in the pathophysiology of diastolic dysfunction, we verified Leprdb/db also displays increased cardiac microvessel thrombosis. Moreover, consistently, with Dhh-deficient mice, we found that aspirin treatment decreased end-diastolic pressure and improved exercise tolerance in Leprdb/db mice. CONCLUSIONS Altogether, these results demonstrate that microvessel thrombosis may participate in the pathophysiology of heart failure with preserved ejection fraction.
Collapse
Affiliation(s)
- Paul Rouault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Sarah Guimbal
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Lauriane Cornuault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Célia Bourguignon
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Ninon Foussard
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Philippe Alzieu
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Frank Choveau
- INSERM U1045, CRCTB (Centre de recherche cardio-thoracique de Bordeaux), IHU Liryc (Institut Hospitalo Universitaire des maladies du rythme cardiaque), University of Bordeaux, France (F.C., D.B.)
| | - David Benoist
- INSERM U1045, CRCTB (Centre de recherche cardio-thoracique de Bordeaux), IHU Liryc (Institut Hospitalo Universitaire des maladies du rythme cardiaque), University of Bordeaux, France (F.C., D.B.)
| | - Candice Chapouly
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Alain-Pierre Gadeau
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Thierry Couffinhal
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| | - Marie-Ange Renault
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1034, Biology of Cardiovascular Diseases, University of Bordeaux, Pessac, France (P.R., S.G., L.C., C.B., N.F., P.A., C.C., A.-P.G., T.C., M.-A.R.)
| |
Collapse
|
8
|
Mora P, Chapouly C. Astrogliosis in multiple sclerosis and neuro-inflammation: what role for the notch pathway? Front Immunol 2023; 14:1254586. [PMID: 37936690 PMCID: PMC10627009 DOI: 10.3389/fimmu.2023.1254586] [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: 07/07/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
Multiple sclerosis is an autoimmune inflammatory disease of the central nervous system leading to neurodegeneration. It affects 2.3 million people worldwide, generally younger than 50. There is no known cure for the disease, and current treatment options - mainly immunotherapies to limit disease progression - are few and associated with serious side effects. In multiple sclerosis, disruption of the blood-brain barrier is an early event in the pathogenesis of lesions, predisposing to edema, excito-toxicity and inflammatory infiltration into the central nervous system. Recently, the vision of the blood brain barrier structure and integrity has changed and include contributions from all components of the neurovascular unit, among which astrocytes. During neuro-inflammation, astrocytes become reactive. They undergo morphological and molecular changes named "astrogliosis" driving the conversion from acute inflammatory injury to a chronic neurodegenerative state. Astrogliosis mechanisms are minimally explored despite their significance in regulating the autoimmune response during multiple sclerosis. Therefore, in this review, we take stock of the state of knowledge regarding astrogliosis in neuro-inflammation and highlight the central role of NOTCH signaling in the process of astrocyte reactivity. Indeed, a very detailed nomenclature published in nature neurosciences in 2021, listing all the reactive astrocyte markers fully identified in the literature, doesn't cover the NOTCH signaling. Hence, we discuss evidence supporting NOTCH1 receptor as a central regulator of astrogliosis in the pathophysiology of neuro-inflammation, notably multiple sclerosis, in human and experimental models.
Collapse
Affiliation(s)
- Pierre Mora
- Université de Bordeaux, Institut national de la santé et de la recherche médicale (INSERM), Biology of Cardiovascular Diseases, Pessac, France
| | | |
Collapse
|
9
|
Panchenko PE, Hippauf L, Konsman JP, Badaut J. Do astrocytes act as immune cells after pediatric TBI? Neurobiol Dis 2023; 185:106231. [PMID: 37468048 PMCID: PMC10530000 DOI: 10.1016/j.nbd.2023.106231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/28/2023] [Accepted: 07/15/2023] [Indexed: 07/21/2023] Open
Abstract
Astrocytes are in contact with the vasculature, neurons, oligodendrocytes and microglia, forming a local network with various functions critical for brain homeostasis. One of the primary responders to brain injury are astrocytes as they detect neuronal and vascular damage, change their phenotype with morphological, proteomic and transcriptomic transformations for an adaptive response. The role of astrocytic responses in brain dysfunction is not fully elucidated in adult, and even less described in the developing brain. Children are vulnerable to traumatic brain injury (TBI), which represents a leading cause of death and disability in the pediatric population. Pediatric brain trauma, even with mild severity, can lead to long-term health complications, such as cognitive impairments, emotional disorders and social dysfunction later in life. To date, the underlying pathophysiology is still not fully understood. In this review, we focus on the astrocytic response in pediatric TBI and propose a potential immune role of the astrocyte in response to trauma. We discuss the contribution of astrocytes in the local inflammatory cascades and secretion of various immunomodulatory factors involved in the recruitment of local microglial cells and peripheral immune cells through cerebral blood vessels. Taken together, we propose that early changes in the astrocytic phenotype can alter normal development of the brain, with long-term consequences on neurological outcomes, as described in preclinical models and patients.
Collapse
Affiliation(s)
| | - Lea Hippauf
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France
| | | | - Jerome Badaut
- CNRS UMR 5536 RMSB-University of Bordeaux, Bordeaux, France; Department of Basic Sciences, Loma Linda University School of Medicine, Loma Linda, CA, USA.
| |
Collapse
|
10
|
Dubey S, Ghosh R, Dubey MJ, Das S, Chakraborty AP, Santra A, Dutta A, Roy D, Pandit A, Roy BK, Das G, Benito-León J. Psychosocial Basis of Human Sufferings and Poverty in Patients with Neurological and Psychiatric Disorders. MEDICAL RESEARCH ARCHIVES 2023; 11:3919. [PMID: 37641666 PMCID: PMC10461571 DOI: 10.18103/mra.v11i5.3919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Neurological disorders and psychiatric ailments often lead to cognitive disabilities and low attainment of education, pivoting misconceptions, myths, and misbeliefs. Poverty and low educational attainment are intriguingly associated with poor awareness and perception of these diseases that add to the suffering. Poverty goes parallel with a low level of education and is intricately associated with neuropsychiatric ailments, which have the potential to spread transgenerationally. Robust education policies, proper government rules and regulations against the spread of disease-related myths and misconceptions, uplifting medical education in its true sense, voices against consanguinity, and programs to raise scientific perception about diseases can help to throw light at the end of this dark tunnel. In this article, the authors intend to 1) decipher the potential psychosocial basis of human suffering and poverty in patients with neurological and psychiatric disorders, and 2) discuss the apropos way-outs that would potentially mitigate suffering, and alleviate the economic burden and cognitive disabilities of families with neuropsychiatric diseases.
Collapse
Affiliation(s)
- Souvik Dubey
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Ritwik Ghosh
- Department of General Medicine, Burdwan Medical College, and Hospital, Burdwan, West Bengal, India
| | - Mahua Jana Dubey
- Department of Psychiatry, Berhampur Mental Hospital, Berhampur, West Bengal, India
| | - Shambaditya Das
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Arka Prava Chakraborty
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Arindam Santra
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Ajitava Dutta
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Dipayan Roy
- Indian Institute of Technology (IIT) Madras, Chennai, Tamil Nadu, India
- School of Humanities, Indira Gandhi National Open University (IGNOU), New Delhi, India
| | - Alak Pandit
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Biman Kanti Roy
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Gautam Das
- Department of Neuromedicine, Bangur Institute of Neurosciences (BIN), Kolkata, West Bengal, India
| | - Julián Benito-León
- Department of Neurology, University Hospital “12 de Octubre”, Madrid, Spain
- Research Institute (i+12), University Hospital “12 de Octubre”, Madrid, Spain
- Centro de Investigación Biomédica en Red Sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
- Department of Medicine, Complutense University, Madrid, Spain
| |
Collapse
|
11
|
Zhou J, Guo P, Duan M, Li J, Ru X, Li L, Guo Z, Zhang JH, Feng H, Chen Y, Sun X. EphA4/EphrinB2 signaling mediates pericyte-induced transient glia limitans formation as a secondary protective barrier after subarachnoid hemorrhage in mice. Exp Neurol 2023; 360:114293. [PMID: 36493862 PMCID: PMC10561606 DOI: 10.1016/j.expneurol.2022.114293] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/13/2022] [Accepted: 12/04/2022] [Indexed: 12/12/2022]
Abstract
BACKGROUND Most patients with subarachnoid hemorrhage (SAH) do not exhibit brain parenchymal injury upon imaging but present significant blood-brain barrier (BBB) disruption and secondary neurological deficits. The aim of this study was to investigate whether stressed astrocytes act as a secondary barrier to exert a protective effect after SAH and to investigate the mechanism of glial limitan formation. METHODS A total of 204 adult male C57BL/6 mice and an endovascular perforation SAH model were employed. The spatiotemporal characteristics of glial limitan formation after SAH were determined by immunofluorescence staining and transmission electron microscopy. The molecular mechanisms by which pericytes regulate glia limitans formation were analyzed using polymerase chain reaction, Western blotting, immunofluorescence staining and ELISA in a pericyte-astrocyte contact coculture system. The findings were validated ex vivo and in vivo using lentiviruses and inhibitors. Finally, pericytes were targeted to regulate glial limitan formation, and the effect of the glia limitans on secondary brain injury after SAH was evaluated by flow cytometry and analysis of neurological function. RESULTS Stress-induced glial limitan formation occurred 1 day after SAH and markedly subsided 3 days after ictus. Pericytes regulated astrocyte glia limitan formation via EphA4/EphrinB2 signaling, inhibited inflammatory cell infiltration and altered neurological function. CONCLUSIONS Astrocyte-derived glia limitans serve as a secondary protective barrier following BBB disruption after SAH in mice, and pericytes can regulate glial limitan formation and alter neurological function via EphA4/EphrinB2 signaling. Strategies for maintaining this secondary protective barrier may be novel treatment approaches for alleviating early brain injury after SAH.
Collapse
Affiliation(s)
- Jiru Zhou
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Peiwen Guo
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Mingxu Duan
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Junhan Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - Xufang Ru
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Lin Li
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China; Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Zongduo Guo
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China
| | - John H Zhang
- Neuroscience Research Center, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States; Department of Physiology and Pharmacology, Loma Linda University School of Medicine, Loma Linda, CA 92354, United States
| | - Hua Feng
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yujie Chen
- Department of Neurosurgery and State Key Laboratory of Trauma, Burn and Combined Injury, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Key Laboratory of Precision Neuromedicine and Neuroregenaration, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China; Chongqing Clinical Research Center for Neurosurgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China.
| | - Xiaochuan Sun
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400016, China.
| |
Collapse
|
12
|
Sousa JA, Bernardes C, Bernardo-Castro S, Lino M, Albino I, Ferreira L, Brás J, Guerreiro R, Tábuas-Pereira M, Baldeiras I, Santana I, Sargento-Freitas J. Reconsidering the role of blood-brain barrier in Alzheimer's disease: From delivery to target. Front Aging Neurosci 2023; 15:1102809. [PMID: 36875694 PMCID: PMC9978015 DOI: 10.3389/fnagi.2023.1102809] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/30/2023] [Indexed: 02/18/2023] Open
Abstract
The existence of a selective blood-brain barrier (BBB) and neurovascular coupling are two unique central nervous system vasculature features that result in an intimate relationship between neurons, glia, and blood vessels. This leads to a significant pathophysiological overlap between neurodegenerative and cerebrovascular diseases. Alzheimer's disease (AD) is the most prevalent neurodegenerative disease whose pathogenesis is still to be unveiled but has mostly been explored under the light of the amyloid-cascade hypothesis. Either as a trigger, bystander, or consequence of neurodegeneration, vascular dysfunction is an early component of the pathological conundrum of AD. The anatomical and functional substrate of this neurovascular degeneration is the BBB, a dynamic and semi-permeable interface between blood and the central nervous system that has consistently been shown to be defective. Several molecular and genetic changes have been demonstrated to mediate vascular dysfunction and BBB disruption in AD. The isoform ε4 of Apolipoprotein E is at the same time the strongest genetic risk factor for AD and a known promoter of BBB dysfunction. Low-density lipoprotein receptor-related protein 1 (LRP-1), P-glycoprotein, and receptor for advanced glycation end products (RAGE) are examples of BBB transporters implicated in its pathogenesis due to their role in the trafficking of amyloid-β. This disease is currently devoid of strategies that change the natural course of this burdening illness. This unsuccess may partly be explained by our misunderstanding of the disease pathogenesis and our inability to develop drugs that are effectively delivered to the brain. BBB may represent a therapeutic opportunity as a target itself or as a therapeutic vehicle. In this review, we aim to explore the role of BBB in the pathogenesis of AD including the genetic background and detail how it can be targeted in future therapeutic research.
Collapse
Affiliation(s)
- João André Sousa
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Catarina Bernardes
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal
| | - Sara Bernardo-Castro
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Miguel Lino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Inês Albino
- Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Lino Ferreira
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - José Brás
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Rita Guerreiro
- Department of Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI, United States
| | - Miguel Tábuas-Pereira
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Inês Baldeiras
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - Isabel Santana
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| | - João Sargento-Freitas
- Department of Neurology, Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal.,Faculty of Medicine, University of Coimbra, Coimbra, Portugal.,Centre for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.,Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
| |
Collapse
|
13
|
Reinhold AK, Salvador E, Förster CY, Birklein F, Rittner HL. Microvascular Barrier Protection by microRNA-183 via FoxO1 Repression: A Pathway Disturbed in Neuropathy and Complex Regional Pain Syndrome. THE JOURNAL OF PAIN 2022; 23:967-980. [PMID: 34974173 DOI: 10.1016/j.jpain.2021.12.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 06/14/2023]
Abstract
Blood nerve barrier disruption and edema are common in neuropathic pain as well as in complex regional pain syndrome (CRPS). MicroRNAs (miRNA) are epigenetic multitarget switches controlling neuronal and non-neuronal cells in pain. The miR-183 complex attenuates hyperexcitability in nociceptors, but additional non-neuronal effects via transcription factors could contribute as well. This study explored exosomal miR-183 in CRPS and murine neuropathy, its effect on the microvascular barrier via transcription factor FoxO1 and tight junction protein claudin-5, and its antihyperalgesic potential. Sciatic miR-183 decreased after CCI. Substitution with perineural miR-183 mimic attenuated mechanical hypersensitivity and restored blood nerve barrier function. In vitro, serum from CCI mice und CRPS patients weakened the microvascular barrier of murine cerebellar endothelial cells, increased active FoxO1 and reduced claudin-5, concomitant with a lack of exosomal miR-183 in CRPS patients. Cellular stress also compromised the microvascular barrier which was rescued either by miR-183 mimic via FoxO1 repression or by prior silencing of Foxo1. PERSPECTIVE: Low miR-183 leading to barrier impairment via FoxO1 and subsequent claudin-5 suppression is a new aspect in the pathophysiology of CRPS and neuropathic pain. This pathway might help untangle the wide symptomatic range of CRPS and nurture further research into miRNA mimics or FoxO1 inhibitors.
Collapse
Affiliation(s)
- Ann-Kristin Reinhold
- University Hospital Würzburg, Department of Anesthesiology, Intensive Care, Emergency Care and Pain Management, Center for Interdisciplinary Pain Medicine, Würzburg, Germany
| | - Ellaine Salvador
- University Hospital Würzburg, Department of Anesthesiology, Intensive Care, Emergency Care and Pain Management, Center for Interdisciplinary Pain Medicine, Würzburg, Germany; University Hospital Würzburg, Department of Neurosurgery, Tumorbiology Laboratory, Würzburg, Germany
| | - Carola Y Förster
- University Hospital Würzburg, Department of Anesthesiology, Intensive Care, Emergency Care and Pain Management, Center for Interdisciplinary Pain Medicine, Würzburg, Germany
| | - Frank Birklein
- Mainz University Hospitals, Department of Neurology, Mainz, Germany
| | - Heike L Rittner
- University Hospital Würzburg, Department of Anesthesiology, Intensive Care, Emergency Care and Pain Management, Center for Interdisciplinary Pain Medicine, Würzburg, Germany.
| |
Collapse
|
14
|
Mou Y, Du Y, Zhou L, Yue J, Hu X, Liu Y, Chen S, Lin X, Zhang G, Xiao H, Dong B. Gut Microbiota Interact With the Brain Through Systemic Chronic Inflammation: Implications on Neuroinflammation, Neurodegeneration, and Aging. Front Immunol 2022; 13:796288. [PMID: 35464431 PMCID: PMC9021448 DOI: 10.3389/fimmu.2022.796288] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Accepted: 02/22/2022] [Indexed: 02/05/2023] Open
Abstract
It has been noticed in recent years that the unfavorable effects of the gut microbiota could exhaust host vigor and life, yet knowledge and theory are just beginning to be established. Increasing documentation suggests that the microbiota-gut-brain axis not only impacts brain cognition and psychiatric symptoms but also precipitates neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). How the blood-brain barrier (BBB), a machinery protecting the central nervous system (CNS) from the systemic circulation, allows the risky factors derived from the gut to be translocated into the brain seems paradoxical. For the unique anatomical, histological, and immunological properties underpinning its permeable dynamics, the BBB has been regarded as a biomarker associated with neural pathogenesis. The BBB permeability of mice and rats caused by GM dysbiosis raises the question of how the GM and its metabolites change BBB permeability and causes the brain pathophysiology of neuroinflammation and neurodegeneration (NF&ND) and brain aging, a pivotal multidisciplinary field tightly associated with immune and chronic systemic inflammation. If not all, gut microbiota-induced systemic chronic inflammation (GM-SCI) mainly refers to excessive gut inflammation caused by gut mucosal immunity dysregulation, which is often influenced by dietary components and age, is produced at the interface of the intestinal barrier (IB) or exacerbated after IB disruption, initiates various common chronic diseases along its dispersal routes, and eventually impairs BBB integrity to cause NF&ND and brain aging. To illustrate the immune roles of the BBB in pathophysiology affected by inflammatory or "leaky" IB resulting from GM and their metabolites, we reviewed the selected publications, including the role of the BBB as the immune barrier, systemic chronic inflammation and inflammation influences on BBB permeability, NF&ND, and brain aging. To add depth to the bridging role of systemic chronic inflammation, a plausible mechanism indispensable for BBB corruption was highlighted; namely, BBB maintenance cues are affected by inflammatory cytokines, which may help to understand how GM and its metabolites play a major role in NF&ND and aging.
Collapse
Affiliation(s)
- Yi Mou
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Yu Du
- Department of Emergency and Critical Care Medicine, The Fourth West China Hospital, Sichuan University, Chengdu, China
| | - Lixing Zhou
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Jirong Yue
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xianliang Hu
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Yixin Liu
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Sao Chen
- Geroscience and Chronic Disease Department, The Eighth Municipal Hospital for the People, Chengdu, China
| | - Xiufang Lin
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Gongchang Zhang
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hengyi Xiao
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Birong Dong
- National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Gueniot F, Rubin S, Bougaran P, Abelanet A, Morel JL, Bontempi B, Proust C, Dufourcq P, Couffinhal T, Duplàa C. Targeting Pdzrn3 maintains adult blood-brain barrier and central nervous system homeostasis. J Cereb Blood Flow Metab 2022; 42:613-629. [PMID: 34644209 PMCID: PMC9051145 DOI: 10.1177/0271678x211048981] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Blood brain barrier (BBB) disruption is a critical component of the pathophysiology of cognitive impairment of vascular etiology (VCI) and associated with Alzheimer's disease (AD). The Wnt pathway plays a crucial role in BBB maintenance, but there is limited data on its role in cognitive pathologies. The E3 ubiquitin ligase PDZRN3 is a regulator of the Wnt pathway. In a murine model of VCI, overexpressing Pdzrn3 in endothelial cell (EC) exacerbated BBB hyperpermeability and accelerated cognitive decline. We extended these observations, in both VCI and AD models, showing that EC-specific depletion of Pdzrn3, reinforced the BBB, with a decrease in vascular permeability and a subsequent spare in cognitive decline. We found that in cerebral vessels, Pdzrn3 depletion protects against AD-induced Wnt target gene alterations and enhances endothelial tight junctional proteins. Our results provide evidence that Wnt signaling could be a molecular link regulating BBB integrity and cognitive decline under VCI and AD pathologies.
Collapse
Affiliation(s)
- Florian Gueniot
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Sebastien Rubin
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Pauline Bougaran
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Alice Abelanet
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | | | | | - Carole Proust
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| | - Pascale Dufourcq
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France.,Service de Biochimie clinique, CHU de Bordeaux, Bordeaux, France
| | - Thierry Couffinhal
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France.,Service des Maladies cardiaques et vasculaires, CHU de Bordeaux, Bordeaux, France
| | - Cecile Duplàa
- Biology of Cardiovascular Diseases, University of Bordeaux U1034, Bordeaux, France
| |
Collapse
|
16
|
Loss-of-function manipulations to identify roles of diverse glia and stromal cells during CNS scar formation. Cell Tissue Res 2022; 387:337-350. [PMID: 34164732 PMCID: PMC8975763 DOI: 10.1007/s00441-021-03487-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Accepted: 06/09/2021] [Indexed: 01/30/2023]
Abstract
Scar formation is the replacement of parenchymal cells by stromal cells and fibrotic extracellular matrix. Until as recently as 25 years ago, little was known about the major functional contributions of different neural and non-neural cell types in the formation of scar tissue and tissue fibrosis in the CNS. Concepts about CNS scar formation are evolving rapidly with the availability of different types of loss-of-function technologies that allow mechanistic probing of cellular and molecular functions in models of CNS disorders in vivo. Such loss-of-function studies are beginning to reveal that scar formation and tissue fibrosis in the CNS involves complex interactions amongst multiple types of CNS glia and non-neural stromal cells. For example, attenuating functions of the CNS resident glial cells, astrocytes or microglia, can disrupt the formation of limitans borders that form around stromal cell scars, which leads to increased spread of inflammation, increased loss of neural tissue, and increased fibrosis. Insights are being gained into specific neuropathological mechanisms whereby specific dysfunctions of different types of CNS glia could cause or contribute to disorder-related tissue pathology and dysfunction. CNS glia, as well as fibrosis-producing stromal cells, are emerging as potential major contributors to diverse CNS disorders either through loss- or gain-of-functions, and are thereby emerging as important potential targets for interventions. In this article, we will review and discuss the effects on CNS scar formation and tissue repair of loss-of-function studies targeted at different specific cell types in various disorder models in vivo.
Collapse
|
17
|
Jansen JA, Omuro A, Lucca LE. T cell dysfunction in glioblastoma: a barrier and an opportunity for the development of successful immunotherapies. Curr Opin Neurol 2021; 34:827-833. [PMID: 34569985 PMCID: PMC8595795 DOI: 10.1097/wco.0000000000000988] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
PURPOSE OF REVIEW Immunotherapies such as immune checkpoint blockade have revolutionized cancer treatment, but current approaches have failed to improve outcomes in glioblastoma and other brain tumours. T cell dysfunction has emerged as one of the major barriers for the development of central nervous system (CNS)-directed immunotherapy. Here, we explore the unique requirements that T cells must fulfil to ensure immune surveillance in the CNS, and we analyse T cell dysfunction in glioblastoma (GBM) through the prism of CNS-resident immune responses. RECENT FINDINGS Using comprehensive and unbiased techniques such as single-cell RNA sequencing, multiple studies have dissected the transcriptional state of CNS-resident T cells that patrol the homeostatic brain. A similar approach has revealed that in GBM, tumour-infiltrating T cells lack the hallmarks of antigen-driven exhaustion typical of melanoma and other solid tumours, suggesting the need for better presentation of tumour-derived antigens. Consistently, in a mouse model of GBM, increasing lymphatic drainage to the cervical lymph node was sufficient to promote tumour rejection. SUMMARY For the success of future immunotherapy strategies, further work needs to explore the natural history of dysfunction in GBM tumour-infiltrating T cells, establish whether these originate from CNS-resident T cells and how they can be manipulated therapeutically.
Collapse
Affiliation(s)
- Josephina A. Jansen
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, USA
| | | | - Liliana E. Lucca
- Departments of Neurology and Immunobiology, Yale School of Medicine, New Haven, USA
| |
Collapse
|
18
|
Hashimoto Y, Campbell M, Tachibana K, Okada Y, Kondoh M. Claudin-5: A Pharmacological Target to Modify the Permeability of the Blood-Brain Barrier. Biol Pharm Bull 2021; 44:1380-1390. [PMID: 34602546 DOI: 10.1248/bpb.b21-00408] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Claudin-5 is the dominant tight junction protein in brain endothelial cells and exclusively limits the paracellular permeability of molecules larger than 400 Da across the blood-brain barrier (BBB). Its pathological impairment or sustained down-regulation has been shown to lead to the progression of psychiatric and neurological disorders, whereas its expression under physiological conditions prevents the passage of drugs across the BBB. While claudin-5 enhancers could potentially act as vascular stabilizers to treat neurological diseases, claudin-5 inhibitors could function as delivery systems to enhance the brain uptake of hydrophilic small-molecular-weight drugs. Therefore, the effects of claudin-5 manipulation on modulating the BBB in different neurological diseases requires further examination. To manipulate claudin-5 expression levels and function, several claudin-5 modulating molecules have been developed. In this review, we first describe the molecular, cellular and pathological aspects of claudin-5 to highlight the mechanisms of claudin-5 enhancers/inhibitors. We then discuss recently developed claudin-5 enhancers/inhibitors and new methods to discover these molecules.
Collapse
Affiliation(s)
| | | | | | - Yoshiaki Okada
- Graduate School of Pharmaceutical Sciences, Osaka University
| | - Masuo Kondoh
- Graduate School of Pharmaceutical Sciences, Osaka University
| |
Collapse
|
19
|
Valori CF, Possenti A, Brambilla L, Rossi D. Challenges and Opportunities of Targeting Astrocytes to Halt Neurodegenerative Disorders. Cells 2021; 10:cells10082019. [PMID: 34440788 PMCID: PMC8395029 DOI: 10.3390/cells10082019] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases are a heterogeneous group of disorders whose incidence is likely to duplicate in the next 30 years along with the progressive aging of the western population. Non-cell-specific therapeutics or therapeutics designed to tackle aberrant pathways within neurons failed to slow down or halt neurodegeneration. Yet, in the last few years, our knowledge of the importance of glial cells to maintain the central nervous system homeostasis in health conditions has increased exponentially, along with our awareness of their fundamental and multifaced role in pathological conditions. Among glial cells, astrocytes emerge as promising therapeutic targets in various neurodegenerative disorders. In this review, we present the latest evidence showing the astonishing level of specialization that astrocytes display to fulfill the demands of their neuronal partners as well as their plasticity upon injury. Then, we discuss the controversies that fuel the current debate on these cells. We tackle evidence of a potential beneficial effect of cell therapy, achieved by transplanting astrocytes or their precursors. Afterwards, we introduce the different strategies proposed to modulate astrocyte functions in neurodegeneration, ranging from lifestyle changes to environmental cues. Finally, we discuss the challenges and the recent advancements to develop astrocyte-specific delivery systems.
Collapse
Affiliation(s)
- Chiara F. Valori
- Molecular Neuropathology of Neurodegenerative Diseases, German Centre for Neurodegenerative Diseases (DZNE), 72076 Tübingen, Germany
- Correspondence: (C.F.V.); (D.R.); Tel.: +49-7071-9254-122 (C.F.V.); +39-0382-592064 (D.R.)
| | - Agostino Possenti
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (A.P.); (L.B.)
| | - Liliana Brambilla
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (A.P.); (L.B.)
| | - Daniela Rossi
- Laboratory for Research on Neurodegenerative Disorders, Istituti Clinici Scientifici Maugeri IRCCS, 27100 Pavia, Italy; (A.P.); (L.B.)
- Correspondence: (C.F.V.); (D.R.); Tel.: +49-7071-9254-122 (C.F.V.); +39-0382-592064 (D.R.)
| |
Collapse
|
20
|
Stavropoulos F, Georgiou E, Sargiannidou I, Kleopa KA. Dysregulation of Blood-Brain Barrier and Exacerbated Inflammatory Response in Cx47-Deficient Mice after Induction of EAE. Pharmaceuticals (Basel) 2021; 14:ph14070621. [PMID: 34203192 PMCID: PMC8308522 DOI: 10.3390/ph14070621] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/18/2022] Open
Abstract
Induction of experimental autoimmune encephalomyelitis (EAE), an animal model of multiple sclerosis (MS), in connexin 32 (Cx32) or Cx47 knockout (KO) mice with deficiency in oligodendrocyte gap junctions (GJs) results in a more severe disease course. In particular, Cx47 KO EAE mice experience an earlier EAE onset and more pronounced disease severity, accompanied by dysregulated pro-inflammatory responses preceding the disease manifestations. In this study, analysis of relevant pro-inflammatory cytokines in wild type EAE, Cx32 KO EAE, and Cx47 KO EAE mice revealed altered expression of Vcam-1 preceding EAE [7 days post injection (dpi)], of Ccl2 at the onset of EAE (12 dpi), and of Gm-csf at the peak of EAE (24 dpi) in Cx47 KO EAE mice. Moreover, Cx47 KO EAE mice exhibited more severe blood-spinal cord barrier (BSCB) disruption, enhanced astrogliosis with defects in tight junction formation at the glia limitans, and increased T-cell infiltration prior to disease onset. Thus, Cx47 deficiency appears to cause dysregulation of the inflammatory profile and BSCB integrity, promoting early astrocyte responses in Cx47 KO EAE mice that lead to a more severe EAE outcome. Further investigation into the role of oligodendrocytic Cx47 in EAE and multiple sclerosis pathology is warranted.
Collapse
Affiliation(s)
- Filippos Stavropoulos
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (F.S.); (E.G.); (I.S.)
| | - Elena Georgiou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (F.S.); (E.G.); (I.S.)
| | - Irene Sargiannidou
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (F.S.); (E.G.); (I.S.)
| | - Kleopas A. Kleopa
- Neuroscience Department, The Cyprus Institute of Neurology and Genetics, Nicosia 2371, Cyprus; (F.S.); (E.G.); (I.S.)
- Center for Multiple Sclerosis and Related Disorders, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia 2371, Cyprus
- Correspondence: ; Tel.: +357-22-358600; Fax: +357-22-392786
| |
Collapse
|
21
|
New Tricks for an Old (Hedge)Hog: Sonic Hedgehog Regulation of Astrocyte Function. Cells 2021; 10:cells10061353. [PMID: 34070740 PMCID: PMC8228508 DOI: 10.3390/cells10061353] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 05/26/2021] [Accepted: 05/27/2021] [Indexed: 01/12/2023] Open
Abstract
The Sonic hedgehog (Shh) molecular signaling pathway is well established as a key regulator of neurodevelopment. It regulates diverse cellular behaviors, and its functions vary with respect to cell type, region, and developmental stage, reflecting the incredible pleiotropy of this molecular signaling pathway. Although it is best understood for its roles in development, Shh signaling persists into adulthood and is emerging as an important regulator of astrocyte function. Astrocytes play central roles in a broad array of nervous system functions, including synapse formation and function as well as coordination and orchestration of CNS inflammatory responses in pathological states. Neurons are the source of Shh in the adult, suggesting that Shh signaling mediates neuron-astrocyte communication, a novel role for this multifaceted pathway. Multiple roles for Shh signaling in astrocytes are increasingly being identified, including regulation of astrocyte identity, modulation of synaptic organization, and limitation of inflammation. This review discusses these novel roles for Shh signaling in regulating diverse astrocyte functions in the healthy brain and in pathology.
Collapse
|
22
|
Chiba H, Ichikawa-Tomikawa N, Imura T, Sugimoto K. The region-selective regulation of endothelial claudin-5 expression and signaling in brain health and disorders. J Cell Physiol 2021; 236:7134-7143. [PMID: 33694168 DOI: 10.1002/jcp.30357] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 02/14/2021] [Accepted: 02/25/2021] [Indexed: 12/26/2022]
Abstract
The neurovascular unit (NVU) consists of neurons, glial cells, microvascular cells, and extracellular matrix, and is involved in a variety of physiological and pathological processes in the central nervous system (CNS). Within the NVU, the microvascular endothelial cells and pericytes principally contribute to maintaining the integrity of the blood-brain barrier (BBB). Various types of cells are connected to each other in the NVU by diverse cell adhesion molecules, of which claudin-5 (CLDN5) is by far the most abundantly expressed tight-junction protein in brain microvascular endothelial cells and absolutely required for the maintenance of the BBB. This review highlights recent progress in understanding the region-specific regulation and dysregulation of CLDN5 expression in CNS health and disorders. We also discuss how CLDN5 expression is regionally disrupted within the NVU. In addition, we focus on the link between cell adhesion and transcription factor signalings and describe the possible involvement of CLDN5-adhesion signaling in brain health and disorders.
Collapse
Affiliation(s)
- Hideki Chiba
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Naoki Ichikawa-Tomikawa
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan
| | - Tetsuya Imura
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan.,Department of Surgical Pathology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kotaro Sugimoto
- Department of Basic Pathology, Fukushima Medical University School of Medicine, Fukushima, Japan
| |
Collapse
|
23
|
Ivan DC, Walthert S, Berve K, Steudler J, Locatelli G. Dwellers and Trespassers: Mononuclear Phagocytes at the Borders of the Central Nervous System. Front Immunol 2021; 11:609921. [PMID: 33746939 PMCID: PMC7973121 DOI: 10.3389/fimmu.2020.609921] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 12/29/2020] [Indexed: 01/02/2023] Open
Abstract
The central nervous system (CNS) parenchyma is enclosed and protected by a multilayered system of cellular and acellular barriers, functionally separating glia and neurons from peripheral circulation and blood-borne immune cells. Populating these borders as dynamic observers, CNS-resident macrophages contribute to organ homeostasis. Upon autoimmune, traumatic or neurodegenerative inflammation, these phagocytes start playing additional roles as immune regulators contributing to disease evolution. At the same time, pathological CNS conditions drive the migration and recruitment of blood-borne monocyte-derived cells across distinct local gateways. This invasion process drastically increases border complexity and can lead to parenchymal infiltration of blood-borne phagocytes playing a direct role both in damage and in tissue repair. While recent studies and technical advancements have highlighted the extreme heterogeneity of these resident and CNS-invading cells, both the compartment-specific mechanism of invasion and the functional specification of intruding and resident cells remain unclear. This review illustrates the complexity of mononuclear phagocytes at CNS interfaces, indicating how further studies of CNS border dynamics are crucially needed to shed light on local and systemic regulation of CNS functions and dysfunctions.
Collapse
|