1
|
Zhang L, Woltering I, Holzner M, Brandhofer M, Schaefer CC, Bushati G, Ebert S, Yang B, Muenchhoff M, Hellmuth JC, Scherer C, Wichmann C, Effinger D, Hübner M, El Bounkari O, Scheiermann P, Bernhagen J, Hoffmann A. CD74 is a functional MIF receptor on activated CD4 + T cells. Cell Mol Life Sci 2024; 81:296. [PMID: 38992165 PMCID: PMC11335222 DOI: 10.1007/s00018-024-05338-5] [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: 03/26/2024] [Revised: 06/04/2024] [Accepted: 06/27/2024] [Indexed: 07/13/2024]
Abstract
Next to its classical role in MHC II-mediated antigen presentation, CD74 was identified as a high-affinity receptor for macrophage migration inhibitory factor (MIF), a pleiotropic cytokine and major determinant of various acute and chronic inflammatory conditions, cardiovascular diseases and cancer. Recent evidence suggests that CD74 is expressed in T cells, but the functional relevance of this observation is poorly understood. Here, we characterized the regulation of CD74 expression and that of the MIF chemokine receptors during activation of human CD4+ T cells and studied links to MIF-induced T-cell migration, function, and COVID-19 disease stage. MIF receptor profiling of resting primary human CD4+ T cells via flow cytometry revealed high surface expression of CXCR4, while CD74, CXCR2 and ACKR3/CXCR7 were not measurably expressed. However, CD4+ T cells constitutively expressed CD74 intracellularly, which upon T-cell activation was significantly upregulated, post-translationally modified by chondroitin sulfate and could be detected on the cell surface, as determined by flow cytometry, Western blot, immunohistochemistry, and re-analysis of available RNA-sequencing and proteomic data sets. Applying 3D-matrix-based live cell-imaging and receptor pathway-specific inhibitors, we determined a causal involvement of CD74 and CXCR4 in MIF-induced CD4+ T-cell migration. Mechanistically, proximity ligation assay visualized CD74/CXCR4 heterocomplexes on activated CD4+ T cells, which were significantly diminished after MIF treatment, pointing towards a MIF-mediated internalization process. Lastly, in a cohort of 30 COVID-19 patients, CD74 surface expression was found to be significantly upregulated on CD4+ and CD8+ T cells in patients with severe compared to patients with only mild disease course. Together, our study characterizes the MIF receptor network in the course of T-cell activation and reveals CD74 as a novel functional MIF receptor and MHC II-independent activation marker of primary human CD4+ T cells.
Collapse
Affiliation(s)
- Lin Zhang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Iris Woltering
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Mathias Holzner
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Markus Brandhofer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Carl-Christian Schaefer
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Genta Bushati
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Simon Ebert
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Bishan Yang
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Maximilian Muenchhoff
- Max von Pettenkofer Institute and Gene Center, Virology, National Reference Center for Retroviruses, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Johannes C Hellmuth
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Department of Medicine III, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Clemens Scherer
- COVID-19 Registry of the LMU Munich (CORKUM), LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
- Department of Medicine I, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Christian Wichmann
- Division of Transfusion Medicine, Cell Therapeutics and Haemostaseology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - David Effinger
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Max Hübner
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany
- Walter Brendel Centre of Experimental Medicine, Ludwig-Maximilians-Universität (LMU) Munich, Munich, Germany
| | - Omar El Bounkari
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany
| | - Patrick Scheiermann
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany
| | - Jürgen Bernhagen
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- German Centre of Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| | - Adrian Hoffmann
- Division of Vascular Biology, Institute for Stroke and Dementia Research (ISD), LMU University Hospital (LMU Klinikum), Ludwig-Maximilians-Universität (LMU) München, Feodor-Lynen-Straße 17, 81377, Munich, Germany.
- Department of Anaesthesiology, LMU University Hospital, Ludwig-Maximilians-Universität (LMU) Munich, Marchioninistraße 15, 81377, Munich, Germany.
- German Centre of Cardiovascular Research (DZHK), Partner Site Munich Heart Alliance, Munich, Germany.
| |
Collapse
|
2
|
Xuan W, Xie W, Li F, Huang D, Zhu Z, Lin Y, Lu B, Yu W, Li Y, Li P. Dualistic roles and mechanistic insights of macrophage migration inhibitory factor in brain injury and neurodegenerative diseases. J Cereb Blood Flow Metab 2023; 43:341-356. [PMID: 36369735 PMCID: PMC9941868 DOI: 10.1177/0271678x221138412] [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: 06/11/2022] [Revised: 09/21/2022] [Accepted: 10/03/2022] [Indexed: 11/15/2022]
Abstract
Macrophage migration inhibitory factor (MIF) is involved in various immune-mediated pathologies and regulates both innate and adaptive immune reactions, thus being related to several acute and chronic inflammatory diseases such as rheumatoid arthritis, septic shock, and atherosclerosis. Its role in acute and chronic brain pathologies, such as stroke and neurodegenerative diseases, has attracted increasing attention in recent years. In response to stimuli like hypoxia, inflammation or infection, different cell types can rapidly release MIF, including immune cells, endothelial cells, and neuron cells. Notably, clinical data from past decades also suggested a possible link between serum MIF levels and the severity of stroke and the evolving of neurodegenerative diseases. In this review, we summarize the major and recent findings focusing on the mechanisms of MIF modulating functions in brain injury and neurodegenerative diseases, which may provide important therapeutic targets meriting further investigation.
Collapse
Affiliation(s)
- Wei Xuan
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Wanqing Xie
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Fengshi Li
- Department of Neurosurgery, Renji Hospital, Shanghai Jiao Tong
University School of Medicine, Shanghai, China
| | - Dan Huang
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Ziyu Zhu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yuxuan Lin
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Binwei Lu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Weifeng Yu
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Yan Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| | - Peiying Li
- Department of Anesthesiology, Clinical Research Center, Renji
Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai,
China
| |
Collapse
|
3
|
Zhao J, Wang X, Li Q, Lu C, Li S. The relevance of serum macrophage migratory inhibitory factor and cognitive dysfunction in patients with cerebral small vascular disease. Front Aging Neurosci 2023; 15:1083818. [PMID: 36824264 PMCID: PMC9941340 DOI: 10.3389/fnagi.2023.1083818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/18/2023] [Indexed: 02/10/2023] Open
Abstract
Cerebral small vascular disease (CSVD) is a common type of cerebrovascular disease, and an important cause of vascular cognitive impairment (VCI) and stroke. The disease burden is expected to increase further as a result of population aging, an ongoing high prevalence of risk factors (e.g., hypertension), and inadequate management. Due to the poor understanding of pathophysiology in CSVD, there is no effective preventive or therapeutic approach for CSVD. Macrophage migration inhibitory factor (MIF) is a multifunctional cytokine that is related to the occurrence and development of vascular dysfunction diseases. Therefore, MIF may contribute to the pathogenesis of CSVD and VCI. Here, reviewed MIF participation in chronic cerebral ischemia-hypoperfusion and neurodegeneration pathology, including new evidence for CSVD, and its potential role in protection against VCI.
Collapse
Affiliation(s)
- Jianhua Zhao
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China,*Correspondence: Jianhua Zhao,
| | - Xiaoting Wang
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Qiong Li
- Henan Joint International Research Laboratory of Neurorestoratology for Senile Dementia, Henan Key Laboratory of Neurorestoratology, Department of Neurology, First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan, China
| | - Chengbiao Lu
- Sino-UK Joint Laboratory of Brain Function and Injury of Henan Province, Department of Physiology and Neurobiology, Xinxiang Medical University, Xinxiang, China
| | - Shaomin Li
- Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
4
|
Li QL, Tang J, Zhao L, Ruze A, Shan XF, Gao XM. The role of CD74 in cardiovascular disease. Front Cardiovasc Med 2023; 9:1049143. [PMID: 36712241 PMCID: PMC9877307 DOI: 10.3389/fcvm.2022.1049143] [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: 09/20/2022] [Accepted: 12/21/2022] [Indexed: 01/15/2023] Open
Abstract
Leukocyte differentiation antigen 74 (CD74), also known as invariant chain, is a molecular chaperone of major histocompatibility complex class II (MHC II) molecules involved in antigen presentation. CD74 has recently been shown to be a receptor for the macrophage migration inhibitory factor family proteins (MIF/MIF2). Many studies have revealed that CD74 plays an important role in cardiovascular disease. In this review, we summarize the structure and main functions of CD74 and then focus on the recent research progress on the role of CD74 in cardiovascular diseases. In addition, we also discuss potential treatment strategies that target CD74. Our systematic review of the role of CD74 in cardiovascular disease will fill some knowledge gaps in the field.
Collapse
Affiliation(s)
- Qiu-Lin Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Jing Tang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China,Department of Clinical Laboratory, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China
| | - Ling Zhao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Amanguli Ruze
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Xue-Feng Shan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China
| | - Xiao-Ming Gao
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asian, Department of Cardiology, The First Affiliated Hospital of Xinjiang Medical University, Ürümqi, China,Xinjiang Key Laboratory of Medical Animal Model Research, Ürümqi, China,Clinical Medical Research Institute of Xinjiang Medical University, Ürümqi, China,*Correspondence: Xiao-Ming Gao,
| |
Collapse
|
5
|
Wang J, Nan Y, Liu M, Hu K. The Role of CD4 + T Cells in the Immunotherapy of Brain Disease by Secreting Different Cytokines. J Neuroimmune Pharmacol 2022; 17:409-422. [PMID: 36443518 DOI: 10.1007/s11481-022-10056-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
Upon different stimulation, naïve CD4+ T cells differentiate into various subsets of T helper (Th) cells, including Th1, Th2, Th17, and Tregs. They play both protective and pathogenic roles in the central nervous system (CNS) by secreting different cytokines. Failure of the homeostasis of the subgroups in the CNS can result in different brain diseases. Recently, immunotherapy has drawn more and more attention in the therapy of various brain diseases. Here, we describe the role of different CD4+ T cell subsets and their secreted cytokines in various brain diseases, as well as the ways in which by affecting CD4+ T cells in therapy of the CNS diseases. Understanding the role of CD4+ T cells and their secreted cytokines in the immunotherapy of brain disease will provide new targets and therapeutics for the treatment of brain disease. The role of CD4 + T cell subtypes in different diseases and their associated regulatory genes, proteins, and enzymes. CD4 + T cell subtypes play both protective (green) and pathogenic (red) roles in different brain diseases. The immune regulatory effects of CD4 + T cells and their subtypes are promoted or inhibited by different genes, proteins, and enzymes.
Collapse
Affiliation(s)
- Jing Wang
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yunrong Nan
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.,Shanghai Innovation Center of TCM Health Service, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Mei Liu
- Industrial Development Center of Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Kaili Hu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
6
|
Neuroprotective Effect of Macrophage Migration Inhibitory Factor (MIF) in a Mouse Model of Ischemic Stroke. Int J Mol Sci 2022; 23:ijms23136975. [PMID: 35805977 PMCID: PMC9267067 DOI: 10.3390/ijms23136975] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/10/2022] Open
Abstract
The mechanism of the neuroprotective effect of the macrophage migration inhibitory factor (MIF) in vivo is unclear. We investigated whether the MIF promotes neurological recovery in an in vivo mouse model of ischemic stroke. Transient middle cerebral artery occlusion (MCAO) surgery was performed to make ischemic stroke mouse model. Male mice were allocated to a sham vehicle, a sham MIF, a middle cerebral artery occlusion (MCAO) vehicle, and MCAO+MIF groups. Transient MCAO (tMCAO) was performed in the MCAO groups, and the vehicle and the MIF were administered via the intracerebroventricular route. We evaluated the neurological functional scale, the rotarod test, and T2-weighted magnetic resonance imaging. The expression level of the microtubule-associated protein 2 (MAP2), Bcl2, and the brain-derived neurotrophic factor (BDNF) were further measured by Western blot assay. The Garcia test was significantly higher in the MCAO+MIF group than in the MCAO+vehicle group. The MCAO+MIF group exhibited significantly better performance on the rotarod test than the MCAO+vehicle group, which further had a significantly reduced total infarct volume on T2-weighted MRI imaging than the MCAO vehicle group. Expression levels of BDNF, and MAP2 tended to be higher in the MCAO+MIF group than in the MCAO+vehicle group. The MIF exerts a neuroprotective effect in an in vivo ischemic stroke model. The MIF facilitates neurological recovery and protects brain tissue from ischemic injury, indicating a possibility of future novel therapeutic agents for stroke patients.
Collapse
|
7
|
Wang J, Zhang J, Ye Y, Xu Q, Li Y, Feng S, Xiong X, Jian Z, Gu L. Peripheral Organ Injury After Stroke. Front Immunol 2022; 13:901209. [PMID: 35720359 PMCID: PMC9200619 DOI: 10.3389/fimmu.2022.901209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/21/2022] [Indexed: 01/08/2023] Open
Abstract
Stroke is a disease with high incidence, mortality and disability rates. It is also the main cause of adult disability in developed countries. Stroke is often caused by small emboli on the inner wall of the blood vessels supplying the brain, which can lead to arterial embolism, and can also be caused by cerebrovascular or thrombotic bleeding. With the exception of recombinant tissue plasminogen activator (rt-PA), which is a thrombolytic drug used to recanalize the occluded artery, most treatments have been demonstrated to be ineffective. Stroke can also induce peripheral organ damage. Most stroke patients have different degrees of injury to one or more organs, including the lung, heart, kidney, spleen, gastrointestinal tract and so on. In the acute phase of stroke, severe inflammation occurs in the brain, but there is strong immunosuppression in the peripheral organs, which greatly increases the risk of peripheral organ infection and aggravates organ damage. Nonneurological complications of stroke can affect treatment and prognosis, may cause serious short-term and long-term consequences and are associated with prolonged hospitalization and increased mortality. Many of these complications are preventable, and their adverse effects can be effectively mitigated by early detection and appropriate treatment with various medical measures. This article reviews the pathophysiological mechanism, clinical manifestations and treatment of peripheral organ injury after stroke.
Collapse
Affiliation(s)
- Jin Wang
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiehua Zhang
- Department of Stomatology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yingze Ye
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qingxue Xu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yina Li
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| | - Shi Feng
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xiaoxing Xiong
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Anesthesia, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
8
|
Marín-Prida J, Liberato JL, Llópiz-Arzuaga A, Stringhetta-Padovani K, Pavón-Fuentes N, Leopoldino AM, Cruz OG, González IH, Pérez ML, Espuny AC, Santos WFDSD, Uyemura SA, Pardo-Andreu GL, Pentón-Rol G. Novel Insights into the Molecular Mechanisms Involved in the Neuroprotective Effects of C-Phycocyanin Against Brain Ischemia in Rats. Curr Pharm Des 2022; 28:1187-1197. [PMID: 35524676 DOI: 10.2174/1381612828666220506145542] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/16/2022] [Indexed: 11/22/2022]
Abstract
BACKGROUND Ischemic stroke produces a large health impact worldwide, with scarce therapeutic options. OBJECTIVE This study aimed to reveal the role of NADPH oxidase and neuroinflammatory genes on the cerebral anti-ischemic effects of C-Phycocyanin (C-PC), the chief biliprotein of Spirulina platensis. METHODS Rats with either focal cerebral ischemia/reperfusion (I/R) or acute brain hypoperfusion, received C-PC at different doses, or a vehicle, for up to 6 h post-stroke. Neurological, behavioral and histochemical parameters were assessed in I/R rats at 24 h. Cerebral gene expression and hippocampal neuron viability were evaluated in hypoperfused rats at acute (24 h) or chronic phases (30 days), respectively. A molecular docking analysis between NOX2 and C-PC-derived Phycocyanobilin (PCB) was also performed. RESULTS C-PC, obtained with a purity of 4.342, significantly reduced the infarct volume and neurologic deficit in a dose-dependent manner, and improved the exploratory activity of the I/R rats. This biliprotein inhibited NOX2 expression, a crucial NAPDH oxidase isoform in the brain, and the superoxide increase produced by the ischemic event. Moreover, C-PC-derived PCB showed a high binding affinity in silico with NOX2. C-PC downregulated the expression of pro-inflammatory genes (IFN-γ, IL-6, IL-17A, CD74, CCL12) and upregulated immune suppressive genes (Foxp3, IL-4, TGF-β) in hypoperfused brain areas. This compound also decreased chronic neuronal death in the hippocampus of hypoperfused rats. CONCLUSION These results suggest that the inhibition of cerebral NADPH oxidase and the improvement of neuroinflammation are key mechanisms mediating the neuroprotective actions of C-PC against brain ischemia.
Collapse
Affiliation(s)
- Javier Marín-Prida
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana
| | - José Luiz Liberato
- Faculty of Philosophy, Sciences and Literature of Ribeirão Preto, University of São Paulo, Brazil
| | | | - Karina Stringhetta-Padovani
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | | | - Andréia Machado Leopoldino
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | - Mariela León Pérez
- Isotopes Center, Ave. Monumental Km 3.5, San José de Las Lajas, Mayabeque, Cuba
| | - Antoni Camins Espuny
- Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Science, University of Barcelona, Barcelona, Spain.,Institute of Neuroscience, University of Barcelona, Barcelona, Spain.,Biomedical Research Networking Centre in Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | | | - Sergio Akira Uyemura
- Department of Clinical Analyses, Toxicology and Food Sciences, Faculty of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Brazil
| | - Gilberto L Pardo-Andreu
- Center for Research and Biological Evaluations, Institute of Pharmacy and Food, University of Havana, Havana, Cuba
| | - Giselle Pentón-Rol
- Center for Genetic Engineering and Biotechnology, Havana, Cuba.,Latin American School of Medicine, Playa, Havana, Cuba
| |
Collapse
|
9
|
CD74 in Apoptotic Macrophages Is Associated with Inflammation, Plaque Progression and Clinical Manifestations in Human Atherosclerotic Lesions. Metabolites 2022; 12:metabo12010054. [PMID: 35050177 PMCID: PMC8781814 DOI: 10.3390/metabo12010054] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Accepted: 01/04/2022] [Indexed: 02/01/2023] Open
Abstract
The aim of this study was to investigate whether CD74 levels in atherosclerotic lesions are associated with inflammation, apoptosis, plaque severity, and clinical symptoms among patients with carotid atherosclerosis. We further studied whether CD74 expression is associated with apoptosis in macrophages induced by 7ketocholesterol (7keto). Sixty-one carotid samples (39 males and 22 females) were immunostained with macrophages, smooth muscle cells, CD74, ferritin, TUNEL (Terminal deoxynucleotidyl transferase dUTP nick end labeling), and thrombin receptors. Double immunocytochemistry of CD74 and caspase 3 or CD74 and Annexin V was performed on THP-1 macrophages exposed to 7keto. In human carotid plaques, CD74 expression is lesion-dependently increased and is associated with necrotic core formation and plaque rupture, clinical symptoms, macrophage apoptosis, ferritin, and thrombin receptors. CD74 levels were inversely correlated to high-density lipoproteins and statin treatment, and positively correlated to triglycerides. In THP-1 macrophages, 7keto induced a significant increase in levels of CD74, ferritin, and apoptotic cell death. This study suggests that CD74 in apoptotic macrophages is linked to inflammation and thrombosis in progression of human atherosclerotic plaques, lipid metabolism, and clinical manifestation in atherosclerosis. Surface CD74 in apoptotic macrophages and ferritin production induced by oxidized lipids may contribute to inflammation and plaque vulnerability in atherosclerosis.
Collapse
|
10
|
Qiu YM, Zhang CL, Chen AQ, Wang HL, Zhou YF, Li YN, Hu B. Immune Cells in the BBB Disruption After Acute Ischemic Stroke: Targets for Immune Therapy? Front Immunol 2021; 12:678744. [PMID: 34248961 PMCID: PMC8260997 DOI: 10.3389/fimmu.2021.678744] [Citation(s) in RCA: 167] [Impact Index Per Article: 55.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 05/31/2021] [Indexed: 12/15/2022] Open
Abstract
Blood-Brain Barrier (BBB) disruption is an important pathophysiological process of acute ischemic stroke (AIS), resulting in devastating malignant brain edema and hemorrhagic transformation. The rapid activation of immune cells plays a critical role in BBB disruption after ischemic stroke. Infiltrating blood-borne immune cells (neutrophils, monocytes, and T lymphocytes) increase BBB permeability, as they cause microvascular disorder and secrete inflammation-associated molecules. In contrast, they promote BBB repair and angiogenesis in the latter phase of ischemic stroke. The profound immunological effects of cerebral immune cells (microglia, astrocytes, and pericytes) on BBB disruption have been underestimated in ischemic stroke. Post-stroke microglia and astrocytes can adopt both an M1/A1 or M2/A2 phenotype, which influence BBB integrity differently. However, whether pericytes acquire microglia phenotype and exert immunological effects on the BBB remains controversial. Thus, better understanding the inflammatory mechanism underlying BBB disruption can lead to the identification of more promising biological targets to develop treatments that minimize the onset of life-threatening complications and to improve existing treatments in patients. However, early attempts to inhibit the infiltration of circulating immune cells into the brain by blocking adhesion molecules, that were successful in experimental stroke failed in clinical trials. Therefore, new immunoregulatory therapeutic strategies for acute ischemic stroke are desperately warranted. Herein, we highlight the role of circulating and cerebral immune cells in BBB disruption and the crosstalk between them following acute ischemic stroke. Using a robust theoretical background, we discuss potential and effective immunotherapeutic targets to regulate BBB permeability after acute ischemic stroke.
Collapse
Affiliation(s)
| | | | | | | | | | - Ya-nan Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bo Hu
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
11
|
Determining the Optimal Administration Conditions under Which MIF Exerts Neuroprotective Effects by Inducing BDNF Expression and Inhibiting Apoptosis in an In Vitro Stroke Model. Brain Sci 2021; 11:brainsci11020280. [PMID: 33672416 PMCID: PMC7926652 DOI: 10.3390/brainsci11020280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/18/2021] [Accepted: 02/20/2021] [Indexed: 12/20/2022] Open
Abstract
Macrophage migration inhibitory factor (MIF) exerts neuroprotective effects against cerebral ischemia/reperfusion injury by inhibiting neuronal apoptosis and inducing the expression of brain-derived neurotrophic factor (BDNF). However, the optimal administration conditions of MIF are currently unknown. Here, we aimed to identify these conditions in an in vitro model. To determine the optimal concentration of MIF, human neuroblastoma cells were assigned to one of seven groups: control, oxygen and glucose deprivation/reperfusion (OGD/R), and OGD/R with different concentrations (1, 10, 30, 60, and 100 ng/mL) of MIF. Six groups were studied to investigate the optimal administration time: control, OGD/R, and OGD/R with MIF administered at different times (pre-OGD, OGD-treat, post-OGD, and whole-processing). Water-soluble tetrazolium salt-1 assay, Western blot analysis, and immunocytochemistry were used to analyze cell viability and protein expression. We found that 60 ng/mL was the optimal concentration of MIF. However, the effects of administration time were not significant; MIF elicited similar neuroprotective effects regardless of administration time. These findings correlated with the expression of BDNF and apoptosis-related proteins. This study provides detailed information on MIF administration, which offers a foundation for future in vivo studies and translation into novel therapeutic strategies for ischemic stroke.
Collapse
|
12
|
Brown J, Kingsbury C, Lee J, Vandenbark AA, Meza‐Romero R, Offner H, Borlongan CV. Spleen participation in partial MHC class II construct neuroprotection in stroke. CNS Neurosci Ther 2020; 26:663-669. [PMID: 32237074 PMCID: PMC7298973 DOI: 10.1111/cns.13369] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/09/2020] [Accepted: 03/10/2020] [Indexed: 12/12/2022] Open
Abstract
Pathological progression of stroke in the peripheral and central nervous systems (PNS and CNS) is characterized by multiple converging signalling pathways that exacerbate neuroinflammation-mediated secondary cell death. This creates a need for a novel type of immunotherapy capable of simultaneously lowering the synergistic inflammatory responses in the PNS and CNS, specifically the spleen and brain. Previously, we demonstrated that partial major histocompatibility complex (MHC) class II constructs can be administered subcutaneously to promote histological and behavioural effects that alleviate common symptoms found in a murine model of transient stroke. This MHC class II manipulates T cell cytokine expression in both PNS and CNS, resulting in dampened inflammation. In our long-standing efforts towards translational research, we recently demonstrated that a potent next generation mouse-based partial MHC class II construct named DRmQ (DRa1L50Q -mMOG-35-55) similarly induces neuroprotection in stroke rats, replicating the therapeutic effects of the human homolog as DRhQ (DRa1L50Q -human (h)MOG-35-55) in stroke mice. Our preclinical studies showed that DRmQ reduces motor deficits, infarct volume and peri-infarct cell loss by targeting inflammation in this second species. Moreover, we provided mechanistic support in both animal studies that partial MHC class II constructs effectively modulate the spleen, an organ which plays a critical role in modulating secondary cell death. Together, these preclinical studies satisfy testing the constructs in two stroke models, which is a major criterion of the Stroke Therapy Academic Industry Roundtable (STAIR) criteria and a key step in effectively translating this drug to the clinic. Additional translational studies, including dose-response and larger animal models may be warranted to bring MHC class II constructs closer to the clinic.
Collapse
Affiliation(s)
- John Brown
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Chase Kingsbury
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Jea‐Young Lee
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| | - Arthur A. Vandenbark
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Roberto Meza‐Romero
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Halina Offner
- Neuroimmunology Research R&D‐31VA Portland Health Care SystemPortlandORUSA,Department of Neurology and Molecular Microbiology & ImmunologyOregon Health & Science UniversityPortlandORUSA
| | - Cesar V. Borlongan
- Department of Neurosurgery and Brain RepairCenter of Excellence for Aging and Brain RepairUniversity of South Florida College of MedicineTampaFLUSA
| |
Collapse
|
13
|
Krishnan S, Lawrence CB. Old Dog New Tricks; Revisiting How Stroke Modulates the Systemic Immune Landscape. Front Neurol 2019; 10:718. [PMID: 31312180 PMCID: PMC6614437 DOI: 10.3389/fneur.2019.00718] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 06/18/2019] [Indexed: 12/27/2022] Open
Abstract
Infections in the post-acute phase of cerebral ischaemia impede optimal recovery by exacerbating morbidity and mortality. Our review aims to reconcile the increased infection susceptibility of patients post-stroke by consolidating our understanding of compartmentalised alterations to systemic immunity. Mounting evidence has catalogued alterations to numerous immune cell populations but an understanding of the mechanisms of long-range communication between the immune system, nervous system and other organs beyond the involvement of autonomic signalling is lacking. By taking our cues from established and emerging concepts of neuro-immune interactions, immune-mediated inter-organ cross-talk, innate immune training and the role of microbiota-derived signals in central nervous system (CNS) function we will explore mechanisms of how cerebral ischaemia could shape systemic immune function. In this context, we will also discuss a key question: how are immune requirements critical for mediating repair of the ischaemic insult balanced by the need for anti-microbial immunity post-stroke, given that they are mediated by mutually exclusive immune networks? Our reformed understanding of the immune landscape post-stroke and novel mechanisms at play could guide targeted therapeutic interventions and initiate a step-change in the clinical management of these infectious complications post-stroke.
Collapse
Affiliation(s)
- Siddharth Krishnan
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Manchester Collaborative Centre for Inflammation Research, University of Manchester, Manchester, United Kingdom.,Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Catherine B Lawrence
- Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom.,Division of Neuroscience and Experimental Psychology, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
14
|
Vandenbark AA, Meza-Romero R, Benedek G, Offner H. A novel neurotherapeutic for multiple sclerosis, ischemic injury, methamphetamine addiction, and traumatic brain injury. J Neuroinflammation 2019; 16:14. [PMID: 30683115 PMCID: PMC6346590 DOI: 10.1186/s12974-018-1393-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 12/27/2018] [Indexed: 02/08/2023] Open
Abstract
Neurovascular, autoimmune, and traumatic injuries of the central nervous system (CNS) all have in common an initial acute inflammatory response mediated by influx across the blood-brain barrier of activated mononuclear cells followed by chronic and often progressive disability. Although some anti-inflammatory therapies can reduce cellular infiltration into the initial lesions, there are essentially no effective treatments for the progressive phase. We here review the successful treatment of animal models for four separate neuroinflammatory and neurodegenerative CNS conditions using a single partial MHC class II construct called DRa1-hMOG-35-55 or its newest iteration, DRa1(L50Q)-hMOG-35-55 (DRhQ) that can be administered without a need for class II tissue type matching due to the conserved DRα1 moiety of the drug. These constructs antagonize the cognate TCR and bind with high affinity to their cell-bound CD74 receptor on macrophages and dendritic cells, thereby competitively inhibiting downstream signaling and pro-inflammatory effects of macrophage migration inhibitory factor (MIF) and its homolog, d-dopachrome tautomerase (D-DT=MIF-2) that bind to identical residues of CD74 leading to progressive disease. These effects suggest the existence of a common pathogenic mechanism involving a chemokine-driven influx of activated monocytes into the CNS tissue that can be reversed by parenteral injection of the DRa1-MOG-35-55 constructs that also induce anti-inflammatory macrophages and microglia within the CNS. Due to their ability to block this common pathway, these novel drugs appear to be prime candidates for therapy of a wide range of neuroinflammatory and neurodegenerative CNS conditions.
Collapse
Affiliation(s)
- Arthur A Vandenbark
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA. .,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA. .,Department of Molecular Microbiology & Immunology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.
| | - Roberto Meza-Romero
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| | - Gil Benedek
- Present Address: Tissue Typing and Immunogenetics Laboratory, Hadassah Medical Center, Jerusalem, Israel
| | - Halina Offner
- Neuroimmunology Research, R&D-31, VA Portland Health Care System, 3710 SW U.S. Veterans Hospital Rd., Portland, OR, 97239, USA.,Department of Neurology, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA.,Department of Anesthesiology and Perioperative Medicine, Oregon Health & Science University, 3181 SW Sam Jackson Park Rd, Portland, OR, 97239, USA
| |
Collapse
|
15
|
Wang Q, Chen X, Yi D, Song Y, Zhao YH, Luo Q. Expression profile analysis of differentially expressed genes in ruptured intracranial aneurysms: In search of biomarkers. Biochem Biophys Res Commun 2018; 506:548-556. [PMID: 30366668 DOI: 10.1016/j.bbrc.2018.10.117] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 10/19/2018] [Indexed: 01/29/2023]
Abstract
Intracranial aneurysms (IAs) result from the bulging of arterial walls secondary to several factors such as flow, vessel morphology, and genetics. Subarachnoid hemorrhage occurs when such walls rupture, leading to high disability and mortality. Despite numerous investigations pertaining to the relationship between geometric characteristics and IA rupture, only a few have obtained consistent results. This study aimed to further identify the potential genes associated with the pathogenesis of IAs, which may provide novel molecular biomarkers. We downloaded and reanalyzed six datasets, which were divided into four groups. IA walls and blood samples were screened for differentially expressed genes (DEGs); then, functional and pathway enrichment analyses were conducted. In total, 158 common DEGs were identified from Groups 1-3 and 396 genes (187 upregulated and 209 downregulated genes) were differentially expressed in Group 4. The functional analysis revealed that the DEGs were mainly associated with the major histocompatibility complex class II protein complex and antigen processing and presentation. Finally, we identified nine key genes, both in aneurysm tissue samples and blood samples, of which three were mostly associated with the progression and rupture of IAs. Bioinformatics was used to analyze the datasets of the ruptured IAs and identify potential biomarkers, which may provide information for the early detection and treatment of IAs.
Collapse
Affiliation(s)
- Qunhui Wang
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Xuan Chen
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Dazhuang Yi
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Yu Song
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, PR China
| | - Yu-Hao Zhao
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, PR China.
| | - Qi Luo
- Department of Neurosurgery, The First Hospital of Jilin University, Changchun, Jilin, PR China.
| |
Collapse
|
16
|
Abstract
Neurocardiology is an emerging specialty that addresses the interaction between the brain and the heart, that is, the effects of cardiac injury on the brain and the effects of brain injury on the heart. This review article focuses on cardiac dysfunction in the setting of stroke such as ischemic stroke, brain hemorrhage, and subarachnoid hemorrhage. The majority of post-stroke deaths are attributed to neurological damage, and cardiovascular complications are the second leading cause of post-stroke mortality. Accumulating clinical and experimental evidence suggests a causal relationship between brain damage and heart dysfunction. Thus, it is important to determine whether cardiac dysfunction is triggered by stroke, is an unrelated complication, or is the underlying cause of stroke. Stroke-induced cardiac damage may lead to fatality or potentially lifelong cardiac problems (such as heart failure), or to mild and recoverable damage such as neurogenic stress cardiomyopathy and Takotsubo cardiomyopathy. The role of location and lateralization of brain lesions after stroke in brain-heart interaction; clinical biomarkers and manifestations of cardiac complications; and underlying mechanisms of brain-heart interaction after stroke, such as the hypothalamic-pituitary-adrenal axis; catecholamine surge; sympathetic and parasympathetic regulation; microvesicles; microRNAs; gut microbiome, immunoresponse, and systemic inflammation, are discussed.
Collapse
Affiliation(s)
- Zhili Chen
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Poornima Venkat
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Don Seyfried
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Michael Chopp
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Tao Yan
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.)
| | - Jieli Chen
- From the Gerontology and Neurological Institute, Tianjin Medical University General Hospital, China (Z.C., T.Y., J.C.); Department of Neurology, Henry Ford Hospital, Detroit, MI (P.V., D.S., M.C., J.C.); and Department of Physics, Oakland University, Rochester, MI (M.C.).
| |
Collapse
|
17
|
Abstract
To achieve success in developing more effective treatments for stroke, we need a better understanding in all aspects of stroke including prevention, diagnosis, treatment, and post-stroke recovery and complications. The objective of this special issue is to bring to the readership of Neurochemistry International the latest developments and knowledge in a broad spectrum of areas of stroke research in both review and original research articles. Topics include neuroprotective diets, biomarkers used to aid clinical management, neurodegenerative as well as neuroprotective effects of the immune system, potential therapeutic targets, engineered growth factors that promote endogenous neuroregeneration, mechanisms of cerebral small vessel disease, and post stroke epilepsy.
Collapse
|