1
|
Castillo-González J, González-Rey E. Beyond wrecking a wall: revisiting the concept of blood-brain barrier breakdown in ischemic stroke. Neural Regen Res 2025; 20:1944-1956. [PMID: 39254550 PMCID: PMC11691464 DOI: 10.4103/nrr.nrr-d-24-00392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Revised: 06/17/2024] [Accepted: 07/04/2024] [Indexed: 09/11/2024] Open
Abstract
The blood-brain barrier constitutes a dynamic and interactive boundary separating the central nervous system and the peripheral circulation. It tightly modulates the ion transport and nutrient influx, while restricting the entry of harmful factors, and selectively limiting the migration of immune cells, thereby maintaining brain homeostasis. Despite the well-established association between blood-brain barrier disruption and most neurodegenerative/neuroinflammatory diseases, much remains unknown about the factors influencing its physiology and the mechanisms underlying its breakdown. Moreover, the role of blood-brain barrier breakdown in the translational failure underlying therapies for brain disorders is just starting to be understood. This review aims to revisit this concept of "blood-brain barrier breakdown," delving into the most controversial aspects, prevalent challenges, and knowledge gaps concerning the lack of blood-brain barrier integrity. By moving beyond the oversimplistic dichotomy of an "open"/"bad" or a "closed"/"good" barrier, our objective is to provide a more comprehensive insight into blood-brain barrier dynamics, to identify novel targets and/or therapeutic approaches aimed at mitigating blood-brain barrier dysfunction. Furthermore, in this review, we advocate for considering the diverse time- and location-dependent alterations in the blood-brain barrier, which go beyond tight-junction disruption or brain endothelial cell breakdown, illustrated through the dynamics of ischemic stroke as a case study. Through this exploration, we seek to underscore the complexity of blood-brain barrier dysfunction and its implications for the pathogenesis and therapy of brain diseases.
Collapse
Affiliation(s)
- Julia Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada, Spain
| | - Elena González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada, Spain
| |
Collapse
|
2
|
Zhou S, Li Z, Li X, Ye Y, Wang M, Jiang J, Tao L, Wang Y, Tung CT, Chung Y, Kim E, Shen X, Xu X, Xiang X, Xie Q, Zhang J, Wu W, Lin X, Chuong CM, Lei M. Crosstalk between endothelial cells and dermal papilla entails hair regeneration and angiogenesis during aging. J Adv Res 2025; 70:339-353. [PMID: 38718895 PMCID: PMC11976415 DOI: 10.1016/j.jare.2024.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/03/2024] [Accepted: 05/05/2024] [Indexed: 05/19/2024] Open
Abstract
INTRODUCTION Tissues maintain their function through interaction with microenvironment. During aging, both hair follicles and blood vessels (BV) in skin undergo degenerative changes. However, it is elusive whether the changes are due to intrinsic aging changes in hair follicles or blood vessels respectively, or their interactions. OBJECTIVE To explore how hair follicles and blood vessels interact to regulate angiogenesis and hair regeneration during aging. METHODS Single-cell RNA-sequencing (scRNA-seq) analyses were used to identify the declined ability of dermal papilla (DP) and endothelial cells (ECs) during aging. CellChat and CellCall were performed to investigate interaction between DP and ECs. Single-cell metabolism (scMetabolism) analysis and iPATH were applied to analyze downstream metabolites in DP and ECs. Hair-plucking model and mouse cell organoid model were used for functional studies. RESULTS During aging, distance and interaction between DP and ECs are decreased. DP interacts with ECs, with decreased EDN1-EDNRA signaling from ECs to DP and CTF1-IL6ST signaling from DP to ECs during aging. ECs-secreted EDN1 binds to DP-expressed EDNRA which enhances Taurine (TA) metabolism to promote hair regeneration. DP-emitted CTF1 binds to ECs-expressed IL6ST which activates alpha-linolenic acid (ALA) metabolism to promote angiogenesis. Activated EDN1-EDNRA-TA signaling promotes hair regeneration in aged mouse skin and in organoid cultures, and increased CTF1-IL6ST-ALA signaling also promotes angiogenesis in aged mouse skin and organoid cultures. CONCLUSIONS Our finding reveals reciprocal interactions between ECs and DP. ECs releases EDN1 sensed by DP to activate TA metabolism which induces hair regeneration, while DP emits CTF1 signal received by ECs to enhance ALA metabolism which promotes angiogenesis. Our study provides new insights into mutualistic cellular crosstalk between hair follicles and blood vessels, and identifies novel signaling contributing to the interactions of hair follicles and blood vessels in normal and aged skin.
Collapse
Affiliation(s)
- Siyi Zhou
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Zeming Li
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xinzhu Li
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Yuanli Ye
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Mengyue Wang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Jingwei Jiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Lihe Tao
- Zhejiang Keyikaijian Bioengineering Co., Ltd, Beijing 100191, China.
| | - Yan Wang
- Zhejiang Keyikaijian Bioengineering Co., Ltd, Beijing 100191, China.
| | - Chen-Tsen Tung
- Zhejiang Keyikaijian Bioengineering Co., Ltd, Beijing 100191, China.
| | | | - Eunmi Kim
- Caregen Co., Ltd, Gyeonggi-do 14119, Korea.
| | - Xinyu Shen
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xuegang Xu
- Department of Dermatology, The First Hospital of China Medical University NHC Key Laboratory of Immunodermatology (China Medical University), Key Laboratory of Immunodermatology (China Medical University), Shenyang, Liaoning Province 110001, China.
| | - Xiao Xiang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Qiaoli Xie
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Jinwei Zhang
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Wang Wu
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Xia Lin
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| | - Cheng-Ming Chuong
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | - Mingxing Lei
- Key Laboratory of Biorheological Science and Technology of Ministry of Education & 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing 400044, China.
| |
Collapse
|
3
|
Zhang X, Li H, Gu Y, Ping A, Chen J, Zhang Q, Xu Z, Wang J, Tang S, Wang R, Lu J, Lu L, Jin C, Jin Z, Zhang J, Shi L. Repair-associated macrophages increase after early-phase microglia attenuation to promote ischemic stroke recovery. Nat Commun 2025; 16:3089. [PMID: 40164598 PMCID: PMC11958652 DOI: 10.1038/s41467-025-58254-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Accepted: 03/12/2025] [Indexed: 04/02/2025] Open
Abstract
Ischemic stroke recovery involves dynamic interactions between the central nervous system and infiltrating immune cells. Peripheral immune cells compete with resident microglia for spatial niches in the brain, but how modulating this balance affects recovery remains unclear. Here, we use PLX5622 to create spatial niches for peripheral immune cells, altering the competition between infiltrating immune cells and resident microglia in male mice following transient middle cerebral artery occlusion (tMCAO). We find that early-phase microglia attenuation promotes long-term functional recovery. This intervention amplifies a subset of monocyte-derived macrophages (RAMf) with reparative properties, characterized by high expression of GPNMB and CD63, enhanced lipid metabolism, and pro-angiogenic activity. Transplantation of RAMf into stroke-affected mice improves white matter integrity and vascular repair. We identify Mafb as the transcription factor regulating the reparative phenotype of RAMf. These findings highlight strategies to optimize immune cell dynamics for post-stroke rehabilitation.
Collapse
Affiliation(s)
- Xiaotao Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- Department of Breast Surgery, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Huaming Li
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Yichen Gu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - An Ping
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jiarui Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Qia Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Zhouhan Xu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Junjie Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Shenjie Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Rui Wang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianan Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
- State Key Laboratory of Transvascular Implantation Devices, Hangzhou, China
| | - Lingxiao Lu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Chenghao Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Ziyang Jin
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China
| | - Jianmin Zhang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
- Brain Research Institute, Zhejiang University, Hangzhou, Zhejiang, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| | - Ligen Shi
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.
- Clinical Research Center for Neurological Diseases of Zhejiang Province, Hangzhou, China.
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, China.
| |
Collapse
|
4
|
Pawluk H, Woźniak A, Tafelska-Kaczmarek A, Kosinska A, Pawluk M, Sergot K, Grochowalska R, Kołodziejska R. The Role of IL-6 in Ischemic Stroke. Biomolecules 2025; 15:470. [PMID: 40305179 PMCID: PMC12024898 DOI: 10.3390/biom15040470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 05/02/2025] Open
Abstract
The pathophysiology of a stroke is a complex process involving oxidative stress and inflammation. As a result of the actions of reactive oxygen species (ROS), not only does vascular damage occur, but the brain tissue is also damaged. It is a dynamic process, induced by a cellular-molecular immune response, focused on the development of an immediate reaction. During ischemia, inflammatory mediators are released, among which IL-6 plays a particularly important role in the acute phase of a stroke. Recently, a lot of attention has been devoted to this pleiotropic pro-inflammatory cytokine, which enhances the migration of leukocytes and is controlled by chemokines and the expression of adhesion handlers. The impact of IL-6 on the severity of neurological treatment and on patient prognosis in AIS is of interest to many researchers. More and more data indicate that it may be a reliable prognostic factor in strokes.
Collapse
Affiliation(s)
- Hanna Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (M.P.); (R.K.)
| | - Alina Woźniak
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (M.P.); (R.K.)
| | - Agnieszka Tafelska-Kaczmarek
- Department of Organic Chemistry, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100 Toruń, Poland;
| | - Agnieszka Kosinska
- Centre for Languages & International Education, University College London, 26 Bedford Way, London WC1H 0AP, UK;
| | - Mateusz Pawluk
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (M.P.); (R.K.)
| | - Krzysztof Sergot
- Laboratory of Laser Molecular Spectroscopy, Institute of Applied Radiation Chemistry, Faculty of Chemistry, Lodz University of Technology, Wroblewskiego 15, 93-590 Lodz, Poland;
| | - Renata Grochowalska
- Laboratory of Cell Biochemistry and Biology, Department of Biotechnology, Institute of Biological Sciences, Faculty of Biological Sciences, University of Zielona Góra, Prof. Szafran 1, 65-516 Zielona Góra, Poland;
| | - Renata Kołodziejska
- Department of Medical Biology and Biochemistry, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (M.P.); (R.K.)
| |
Collapse
|
5
|
Hall C, Nguyen DT, Mendoza K, Tan C, Chauhan A. Inhibition of IL-6 trans-signaling promotes post-stroke functional recovery in a sex and dose-dependent manner. J Neuroinflammation 2025; 22:52. [PMID: 40011978 PMCID: PMC11866694 DOI: 10.1186/s12974-025-03365-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 02/01/2025] [Indexed: 02/28/2025] Open
Abstract
INTRODUCTION Elevated circulating IL-6 levels are associated with poorer outcomes after stroke, and increased serum IL-6 levels are linked to a higher risk of stroke. IL-6 binds to soluble IL-6 receptors (sIL-6R) and subsequently to ubiquitously expressed gp130, initiating proinflammatory trans-signaling. This study tested the hypothesis that inhibiting IL-6 trans-signaling by administering soluble (s) gp130 improves long-term functional outcomes in young mice after stroke. METHODS Recombinant mouse gp130Fc chimera (sgp130) was administered one hour after middle cerebral artery occlusion (MCAO) followed by twice-weekly administration for 2 weeks in mice (8-15 weeks old). Behavioral assessments were done on days 7 and 28 post-MCAO for chronic studies. Flow cytometry was performed on days 3 (blood) and 7 (spleen and brain) to assess IL-6, mIL-6R, and phosphorylated STAT3 expression. RESULTS Improved long-term functional outcomes were observed in male, but not female mice. To investigate the differential response in females, ELISA analyses revealed that plasma IL-6 levels increased in both sexes after MCAO, with a more pronounced induction in females. Additionally, circulating sIL-6R levels were significantly higher in females compared to males (p < 0.05) at 24 h post-MCAO. Administering a higher dose of sgp130 (1 mg/kg) to females improved long-term functional outcomes, suggesting that a higher dose is needed to inhibit IL-6 trans-signaling in females effectively. Mechanistically, sgp130 treatment reduced phosphorylated STAT3 expression in brain F4/80 macrophages and increased the expression of mIL-6R on splenic immune cells at day 7 post-MCAO in both sexes. CONCLUSION These findings demonstrate that inhibition of IL-6 trans-signaling with gp130Fc improves long-term functional outcomes in both male and female mice, albeit in a dose-dependent manner. This study provides novel insights into potential therapeutic strategies targeting IL-6 signaling pathways following stroke.
Collapse
Affiliation(s)
- Cassandra Hall
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Dustin T Nguyen
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Kate Mendoza
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Chunfeng Tan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA
| | - Anjali Chauhan
- Department of Neurology, University of Texas McGovern Medical School at Houston, Houston, TX, USA.
| |
Collapse
|
6
|
Dargazanli C, Blaquière M, Moynier M, de Bock F, Labreuche J, Ter Schiphorst A, Derraz I, Radu RA, Gascou G, Lefevre PH, Rapido F, Fendeleur J, Arquizan C, Bourcier R, Marin P, Machi P, Cagnazzo F, Hirtz C, Costalat V, Marchi N. Inflammation biomarkers in the intracranial blood are associated with outcome in patients with ischemic stroke. J Neurointerv Surg 2025; 17:159-166. [PMID: 38514190 DOI: 10.1136/jnis-2023-021365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/18/2024] [Indexed: 03/23/2024]
Abstract
BACKGROUND Performing endovascular treatment (EVT) in patients with acute ischemic stroke (AIS) allows a port of entry for intracranial biological sampling. OBJECTIVE To test the hypothesis that specific immune players are molecular contributors to disease, outcome biomarkers, and potential targets for modifying AIS . METHODS We examined 75 subjects presenting with large vessel occlusion of the anterior circulation and undergoing EVT. Intracranial blood samples were obtained by microcatheter aspiration, as positioned for stent deployment. Peripheral blood samples were collected from the femoral artery. Plasma samples were quality controlled by electrophoresis and analyzed using a Mesoscale multiplex for targeted inflammatory and vascular factors. RESULTS We measured 37 protein biomarkers in our sample cohort. Through multivariate analysis, adjusted for age, intravenous thrombolysis, pretreatment National Institutes of Health Stroke Scale and Alberta Stroke Program Early CT scores, we found that post-clot blood levels of interleukin-6 (IL-6) were significantly correlated (adjusted P value <0.05) with disability assessed by the modified Rankin Scale (mRS) score at 90 days, with medium effect size. Chemokine (C-C) ligand 17 CCL17/TARC levels were inversely correlated with the mRS score. Examination of peripheral blood showed that these correlations did not reach statistical significance after correction. Intracranial biomarker IL-6 level was specifically associated with a lower likelihood of favorable outcome, defined as a mRS score of 0-2. CONCLUSIONS Our findings show a signature of blood inflammatory factors at the cerebrovascular occlusion site. The correlations between these acute-stage biomarkers and mRS score outcome support an avenue for add-on and localized immune modulatory strategies in AIS.
Collapse
Affiliation(s)
- Cyril Dargazanli
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Marine Blaquière
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Marinette Moynier
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
| | - Frédéric de Bock
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julien Labreuche
- Unité Statistique, Évaluation Économique, Data-management, Centre Hospitalier Universitaire de Lille, Lille, France
| | - Adrien Ter Schiphorst
- Department of Neurology, CHRU Gui de Chauliac, University Hospital Centre Montpellier, Montpellier, France
| | - Imad Derraz
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
| | - Răzvan Alexandru Radu
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
| | - Gregory Gascou
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
| | - Pierre Henri Lefevre
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
| | - Francesca Rapido
- Department of Anesthesiology and Critical Care Medicine, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Julien Fendeleur
- Department of Anesthesiology and Critical Care Medicine, Gui de Chauliac Hospital, Montpellier University Medical Center, Montpellier, France
| | - Caroline Arquizan
- Department of Neurology, CHRU Gui de Chauliac, University Hospital Centre Montpellier, Montpellier, France
| | - Romain Bourcier
- Department of Neuroradiology, Université de Nantes, Nantes, France
| | - Philippe Marin
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Paolo Machi
- Department of Neuroradiology, Geneva University Hospitals, Geneve, Switzerland
| | - Federico Cagnazzo
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | | | - Vincent Costalat
- Department of Neuroradiology, University Hospital Centre Montpellier, Montpellier, France
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicola Marchi
- Institute of Functional Genomics, University of Montpellier, CNRS, INSERM, Montpellier, France
| |
Collapse
|
7
|
Iadecola C, Anrather J. The immunology of stroke and dementia. Immunity 2025; 58:18-39. [PMID: 39813992 PMCID: PMC11736048 DOI: 10.1016/j.immuni.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Ischemic stroke and vascular cognitive impairment, caused by a sudden arterial occlusion or more subtle but protracted vascular insufficiency, respectively, are leading causes of morbidity and mortality worldwide with limited therapeutic options. Innate and adaptive immunity have long been implicated in neurovascular injury, but recent advances in methodology and new experimental approaches have shed new light on their contributions. A previously unappreciated dynamic interplay of brain-resident, meningeal, and systemic immune cells with the ischemic brain and its vasculature has emerged, and new insights into the frequent overlap between vascular and Alzheimer pathology have been provided. Here, we critically review these recent findings, place them in the context of current concepts on neurovascular pathologies and Alzheimer's disease, and highlight their impact on recent stroke and Alzheimer therapies.
Collapse
Affiliation(s)
- Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY 10065, USA.
| |
Collapse
|
8
|
Castillo-González J, Buscemi L, Vargas-Rodríguez P, Serrano-Martínez I, Forte-Lago I, Caro M, Price M, Hernández-Cortés P, Hirt L, González-Rey E. Cortistatin exerts an immunomodulatory and neuroprotective role in a preclinical model of ischemic stroke. Pharmacol Res 2024; 210:107501. [PMID: 39521024 DOI: 10.1016/j.phrs.2024.107501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/24/2024] [Accepted: 11/06/2024] [Indexed: 11/16/2024]
Abstract
Ischemic stroke is the result of a permanent or transient occlusion of a brain artery, leading to irreversible tissue injury and long-term sequelae. Despite ongoing advancements in revascularization techniques, stroke remains the second leading cause of death worldwide. A comprehensive understanding of the complex and interconnected mechanisms, along with the endogenous mediators that modulate stroke responses is essential for the development of effective interventions. Our study investigates cortistatin, a neuropeptide extensively distributed in the immune and central nervous systems, known for its immunomodulatory properties. With neuroinflammation and peripheral immune deregulation as key pathological features of brain ischemia, cortistatin emerges as a promising therapeutic candidate. To this aim, we evaluated its potential effect in a well-established middle cerebral artery occlusion (MCAO) preclinical stroke model. Our findings indicated that the peripheral administration of cortistatin at 24 h post-stroke significantly reduced neurological damage and enhanced recovery. Importantly, cortistatin-induced neuroprotection was multitargeted, as it modulated the glial reactivity and astrocytic scar formation, facilitated blood-brain barrier recovery, and regulated local and systemic immune dysfunction. Surprisingly, administration of cortistatin at immediate and early post-stroke time points proved to be not beneficial and even detrimental. These results emphasize the importance of understanding the spatio-temporal dynamics of stroke pathology to develop innovative therapeutic strategies with appropriate time windows. Premature interruption of certain neuroinflammatory processes might inadvertently compromise neuroprotective mechanisms. In summary, our study highlights cortistatin as a novel pleiotropic therapeutic approach against ischemic stroke, offering new treatment options for patients who undergo early revascularization intervention but unsuccessful recovery.
Collapse
Affiliation(s)
- J Castillo-González
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada 18016, Spain
| | - L Buscemi
- University of Lausanne, Lausanne, Switzerland; Lausanne University Hospital, Lausanne, Switzerland
| | - P Vargas-Rodríguez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada 18016, Spain
| | - I Serrano-Martínez
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada 18016, Spain
| | - I Forte-Lago
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada 18016, Spain
| | - M Caro
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada 18016, Spain
| | - M Price
- University of Lausanne, Lausanne, Switzerland; Lausanne University Hospital, Lausanne, Switzerland
| | | | - L Hirt
- University of Lausanne, Lausanne, Switzerland; Lausanne University Hospital, Lausanne, Switzerland.
| | - E González-Rey
- Institute of Parasitology and Biomedicine Lopez-Neyra (IPBLN), CSIC, PT Salud, Granada 18016, Spain.
| |
Collapse
|
9
|
Khan H, Naseem T, Kaushik P, Narang J, Khan R, Panwar S, Parvez S. Decoding paradoxical links of cytokine markers in cognition: Cross talk between physiology, inflammaging, and Alzheimer's disease- related cognitive decline. Ageing Res Rev 2024; 101:102535. [PMID: 39374831 DOI: 10.1016/j.arr.2024.102535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 09/29/2024] [Accepted: 10/01/2024] [Indexed: 10/09/2024]
Abstract
Recent research has revolutionized our understanding of memory consolidation by emphasizing the critical role of astrocytes, microglia, and immune cells in through cytokine signaling. Cytokines, compact proteins, play pivotal roles in neuronal development, synaptic transmission, and normal aging. This review explores the cellular mechanisms contributing to cognitive decline in inflammaging and Alzheimer's disease, highlighting the paradoxical effects of most studied cytokines (IL-1, IL-6, TNF-α) in brain function, which act as a double-edged sword in brain physiology, acting both as facilitators of healthy cognitive function and as a potential contributor to cognitive decline.
Collapse
Affiliation(s)
- Hiba Khan
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Talib Naseem
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Pooja Kaushik
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Jagriti Narang
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Rehan Khan
- Chemical Biology Unit, Institute of Nano Science and Technology, Sector 81, Knowledge City, Sahibzada Ajit Singh Nagar, Mohali, Punjab 140306, India
| | - Siddharth Panwar
- School of Computing and Electrical Engineering, Indian Institute of Technology, Mandi, Himachal Pradesh 175075, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
10
|
Yu L, Zhang C, Gu L, Chen H, Huo Y, Wang S, Tao J, Xu C, Zhang Q, Ma M, Zhang J. Hydroxysafflor Yellow A and Tenuigenin Exhibit Neuroprotection Effects Against Focal Cerebral Ischemia Via Differential Regulation of JAK2/STAT3 and SOCS3 Signaling Interaction. Mol Neurobiol 2024; 61:5584-5600. [PMID: 38214838 DOI: 10.1007/s12035-023-03896-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/19/2023] [Indexed: 01/13/2024]
Abstract
Numerous natural bioactive compounds extracted from Chinese medicines have been proved to be promising and potent agents in the treatment of ischemic stroke. Hydroxysafflor yellow A (HSYA), separated from Carthamus tinctorius, has increasingly attracted attention for its broad spectrum of pharmacological effects, especially of its neuroprotective action. Our previous studies revealed that HSYA plays significant beneficial roles in a dose-dependent manner in rats with focal cerebral ischemia. However, treatment with higher doses of HSYA appeared to bring about adverse reactions in the rats. In present study, we adopted tenuigenin (TEN), extracted from the Polygala tenuifolia root, in combination with HSYA to optimize the therapeutic strategy against ischemic stroke, and further explored the underlying mechanisms of action of the combination in vivo and in vitro. We firstly confirmed the pharmacological efficacies of co-treatment of HSYA and TEN in middle cerebral ischemia occlusion (MCAO) rats and observed the synergistic improvement of infarct volume, cerebral edema, and morphology of neuron cell body. Behavioral experiments indicated that combination of HSYA and TEN could synergistically improve motor and cognitive function in MCAO rats. We also observed increased viability and suppressed cell apoptosis after HSYA and TEN co-treatments in the oxygen-glucose deprivation/reperfusion (OGD/R) SH-SY5Y cells. Furthermore, JAK2/STAT3 and SOCS3 signaling interaction was demonstrated to be a critical responsor to the co-treatment of HSYA and TEN. In the subsequent experiments with silencing SOCS3 in OGD/R-exposed cells, we found that HSYA and TEN might suppress JAK2/STAT3 pathway through different regulatory mechanisms targeting SOCS3-negative feedback signaling. HSYA seemed to impose excessive activation of JAK2/STAT3 to trigger SOCS3-negative feedback signaling, while TEN appeared to provoke SOCS3 inhibitory feedback role directly to further attenuate JAK2-mediated signaling. Collectively, HSYA and TEN might modulate the crosstalk between JAK2/STAT3 and SOCS3 signaling pathways in different manners that eventually contributed to their synergistic therapeutic effects against cerebral ischemic stroke.
Collapse
Affiliation(s)
- Lu Yu
- Comprehensive Department of Traditional Chinese Medicine, Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Cheng Zhang
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Lingling Gu
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062, China
| | - Hong Chen
- Department of Clinical Laboratory, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China
| | - Yan Huo
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China
| | - Shuyan Wang
- Department of Anesthesiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China
| | - Jie Tao
- Comprehensive Department of Traditional Chinese Medicine, Department of Neurology, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200062, China
| | - Chuan Xu
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China
| | - Qiujuan Zhang
- Department of Neurology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200437, China.
| | - Mingliang Ma
- Key Laboratory of Brain Functional Genomics, Ministry of Education, School of Life Sciences, East China Normal University, Shanghai, 200062, China.
- Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200062, China.
| | - Jun Zhang
- Department of Clinical Laboratory, Shanghai Key Laboratory of Maternal Fetal Medicine, Shanghai Institute of Maternal-Fetal Medicine and Gynecologic Oncology, Shanghai First Maternity and Infant Hospital, School of Medicine, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
11
|
Benkő S, Dénes Á. Microglial Inflammatory Mechanisms in Stroke: The Jury Is Still Out. Neuroscience 2024; 550:43-52. [PMID: 38364965 DOI: 10.1016/j.neuroscience.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 02/09/2024] [Accepted: 02/12/2024] [Indexed: 02/18/2024]
Abstract
Microglia represent the main immune cell population in the CNS with unique homeostatic roles and contribution to broad neurological conditions. Stroke is associated with marked changes in microglial phenotypes and induction of inflammatory responses, which emerge as key modulators of brain injury, neurological outcome and regeneration. However, due to the limited availability of functional studies with selective targeting of microglia and microglia-related inflammatory pathways in stroke, the vast majority of observations remain correlative and controversial. Because extensive review articles discussing the role of inflammatory mechanisms in different forms of acute brain injury are available, here we focus on some specific pathways that appear to be important for stroke pathophysiology with assumed contribution by microglia. While the growing toolkit for microglia manipulation increasingly allows targeting inflammatory pathways in a cell-specific manner, reconsideration of some effects devoted to microglia may also be required. This may particularly concern the interpretation of inflammatory mechanisms that emerge in response to stroke as a form of sterile injury and change markedly in chronic inflammation and common stroke comorbidities.
Collapse
Affiliation(s)
- Szilvia Benkő
- Laboratory of Inflammation-Physiology, Department of Physiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary.
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest H-1083, Hungary.
| |
Collapse
|
12
|
Carnwath TP, Demel SL, Prestigiacomo CJ. Genetics of ischemic stroke functional outcome. J Neurol 2024; 271:2345-2369. [PMID: 38502340 PMCID: PMC11055934 DOI: 10.1007/s00415-024-12263-x] [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: 11/20/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 03/21/2024]
Abstract
Ischemic stroke, which accounts for 87% of cerebrovascular accidents, is responsible for massive global burden both in terms of economic cost and personal hardship. Many stroke survivors face long-term disability-a phenotype associated with an increasing number of genetic variants. While clinical variables such as stroke severity greatly impact recovery, genetic polymorphisms linked to functional outcome may offer physicians a unique opportunity to deliver personalized care based on their patient's genetic makeup, leading to improved outcomes. A comprehensive catalogue of the variants at play is required for such an approach. In this review, we compile and describe the polymorphisms associated with outcome scores such as modified Rankin Scale and Barthel Index. Our search identified 74 known genetic polymorphisms spread across 48 features associated with various poststroke disability metrics. The known variants span diverse biological systems and are related to inflammation, vascular homeostasis, growth factors, metabolism, the p53 regulatory pathway, and mitochondrial variation. Understanding how these variants influence functional outcome may be helpful in maximizing poststroke recovery.
Collapse
Affiliation(s)
- Troy P Carnwath
- University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA.
| | - Stacie L Demel
- Department of Neurology, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| | - Charles J Prestigiacomo
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, OH, 45267, USA
| |
Collapse
|
13
|
Smit RD, Ghosh B, Campion TJ, Stingel R, Lavell E, Hooper R, Fan X, Soboloff J, Smith GM. STAT3 protects dopaminergic neurons against degeneration in animal model of Parkinson's disease. Brain Res 2024; 1824:148691. [PMID: 38030102 PMCID: PMC10842767 DOI: 10.1016/j.brainres.2023.148691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 11/22/2023] [Accepted: 11/26/2023] [Indexed: 12/01/2023]
Abstract
INTRODUCTION Parkinson's disease (PD) is the most prevalent disorder of the basal ganglia, propagated by the degeneration of axon terminals within the striatum and subsequent loss of dopaminergic neurons in the substantia nigra (SN). Exposure of environmental neurotoxins and mutations of several mitochondrial and proteasomal genes are primarily responsible. METHODS To determine whether signal transducer and activator of transcription 3 (STAT3) could protect dopaminergic neurons against degeneration, we first screened it in the in vitro capacity using immortalized rat dopaminergic N27 cells under 6-OHDA neurotoxicity. We then evaluated the effectiveness of constitutively active (ca) STAT3 as a neuroprotective agent on N27 cells in a 6-hydroxydopamine (6-OHDA) induced rat model of PD and compared it to control animals or animals where AAV/caRheb was expressed in SN. Behavioral outcomes were assessed using rotational and cylinder assays and mitochondrial function using reactive oxygen species (ROS) levels. RESULTS Using flow cytometry, the in vitro analysis determined caSTAT3 significantly decreased dopaminergic neuronal death under 6-OHDA treatment conditions. Importantly, in vivo overexpression of caSTAT3 in SN dopaminergic neurons using AAV-mediated expression demonstrated significant neuroprotection of dopaminergic neurons following 6-OHDA. Both caSTAT3 and caRheb + caSTAT3 co-injection into substantia nigra reduced D-amphetamine-induced rotational behavior and increased ipsilateral forelimb function when compared to control animals. In addition, caSTAT3 decreased mitochondrial ROS production following 6-OHDA induced neurotoxicity. CONCLUSION caSTAT3 confers resistance against ROS production in mitochondria of susceptible SN dopaminergic neurons potentially offering a new avenue for treatment against PD.
Collapse
Affiliation(s)
- Rupert D Smit
- Department of Neuroscience & Shriners Hospitals for Pediatric Research Center, Temple University, USA.
| | - Biswarup Ghosh
- Department of Neuroscience & Shriners Hospitals for Pediatric Research Center, Temple University, USA
| | - Thomas J Campion
- Department of Neuroscience & Shriners Hospitals for Pediatric Research Center, Temple University, USA
| | - Rachel Stingel
- Department of Neuroscience & Shriners Hospitals for Pediatric Research Center, Temple University, USA
| | - Emily Lavell
- Department of Neuroscience & Shriners Hospitals for Pediatric Research Center, Temple University, USA
| | - Robert Hooper
- Fels Institute for Cancer Research & Molecular Biology, Temple University, USA
| | - Xiaoxuan Fan
- Flow Cytometry Core Facility, Temple University, USA
| | - Jonathan Soboloff
- Fels Institute for Cancer Research & Molecular Biology, Temple University, USA
| | - George M Smith
- Department of Neuroscience & Shriners Hospitals for Pediatric Research Center, Temple University, USA
| |
Collapse
|
14
|
Mezuki S, Matsuo R, Irie F, Shono Y, Kuwashiro T, Sugimori H, Wakisaka Y, Ago T, Kamouchi M, Kitazono T, on behalf of the Fukuoka Stroke Registry Investigators. Body temperature in the acute phase and clinical outcomes after acute ischemic stroke. PLoS One 2024; 19:e0296639. [PMID: 38206979 PMCID: PMC10783745 DOI: 10.1371/journal.pone.0296639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/15/2023] [Indexed: 01/13/2024] Open
Abstract
BACKGROUND This study aimed to examine whether post-stroke early body temperature is associated with neurological damage in the acute phase and functional outcomes at three months. METHODS We included 7,177 patients with acute ischemic stroke within 24 h of onset. Axillary temperature was measured daily in the morning for seven days. Mean body temperature was grouped into five quintiles (Q1: 35.1‒36.5°C, Q2: 36.5‒36.7°C, Q3: 36.7‒36.8°C, Q4: 36.8‒37.1°C, and Q5: 37.1‒39.1°C). Clinical outcomes included neurological improvement during hospitalization and poor functional outcome (modified Rankin scale score, 3-6) at three months. A logistic regression analysis was performed to evaluate the association between body temperature and clinical outcomes. RESULTS The patient's mean (SD) age was 70.6 (12.3) years, and 35.7% of patients were women. Mean body temperature was significantly associated with less neurological improvement from Q2 (odds ratios [95% confidence interval], 0.77 [0.65-0.99] vs. Q1) to Q5 (0.33 [0.28-0.40], P for trend <0.001) even after adjusting for potential confounders, including baseline neurological severity, C-reactive protein levels, and post-stroke acute infections. The multivariable-adjusted risk of poor functional outcome linearly increased from Q2 (1.36 [1.03-1.79]) to Q5 (6.44 [5.19-8.96], P for trend <0.001). These associations were maintained even in the analyses excluding patients with acute infectious diseases. Multivariable-adjusted risk of poor functional outcome was higher in patients with early body temperature elevation on days 1-3 and with longer duration with body temperature >37.0°C. CONCLUSIONS Post-stroke early high body temperature is independently associated with unfavorable outcomes following acute ischemic stroke.
Collapse
Affiliation(s)
- Satomi Mezuki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Ryu Matsuo
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Fumi Irie
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Shono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Emergency and Critical Care Center, Kyushu University Hospital, Fukuoka, Japan
| | - Takahiro Kuwashiro
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cerebrovascular Medicine and Neurology, Kyushu Medical Center, Fukuoka, Japan
| | - Hiroshi Sugimori
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Cerebrovascular Medicine and Neurology, Kyushu Medical Center, Fukuoka, Japan
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Masahiro Kamouchi
- Department of Health Care Administration and Management, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
- Center for Cohort Study, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | |
Collapse
|
15
|
Li D, Lian L, Huang L, Gamdzyk M, Huang Y, Doycheva D, Li G, Yu S, Guo Y, Kang R, Tang H, Tang J, Kong L, Zhang JH. Delayed recanalization reduced neuronal apoptosis and neurological deficits by enhancing liver-derived trefoil factor 3-mediated neuroprotection via LINGO2/EGFR/Src signaling pathway after middle cerebral artery occlusion in rats. Exp Neurol 2024; 371:114607. [PMID: 37935323 PMCID: PMC11585322 DOI: 10.1016/j.expneurol.2023.114607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Delayed recanalization at days or weeks beyond the therapeutic window was shown to improve functional outcomes in acute ischemic stroke (AIS) patients. However, the underlying mechanisms remain unclear. Previous preclinical study reported that trefoil factor 3 (TFF3) was secreted by liver after cerebral ischemia and acted a distant neuroprotective factor. Here, we investigated the liver-derived TFF3-mediated neuroprotective mechanism enhanced by delayed recanalization after AIS. A total of 327 male Sprague-Dawley rats and the model of middle cerebral artery occlusion (MCAO) with permanent occlusion (pMCAO) or with delayed recanalization at 3 d post-occlusion (rMCAO) were used. Partial hepatectomy was performed within 5 min after MCAO. Leucine-rich repeat and immunoglobulin-like domain-containing nogo receptor-interacting protein 2 (LINGO2) siRNA was administered intracerebroventricularly at 48 h after MCAO. Recombinant rat TFF3 (rr-TFF3, 30 μg/Kg) or recombinant rat epidermal growth factor (rr-EGF, 100 μg/Kg) was administered intranasally at 1 h after recanalization, and EGFR inhibitor Gefitinib (75 mg/Kg) was administered intranasally at 30 min before recanalization. The evaluation of outcomes included neurobehavior, ELISA, western blot and immunofluorescence staining. TFF3 in hepatocytes and serum were upregulated in a similar time-dependent manner after MCAO. Compared to pMCAO, delayed recanalization increased brain TFF3 levels and attenuated brain damage with the reduction in neuronal apoptosis, infarct volume and neurological deficits. Partial hepatectomy reduced TFF3 levels in serum and ipsilateral brain hemisphere, and abolished the benefits of delayed recanalization on neuronal apoptosis and neurobehavioral deficits in rMCAO rats. Intranasal rrTFF3 treatment reversed the changes associated with partial hepatectomy. Delayed recanalization after MCAO increased the co-immunoprecipitation of TFF3 and LINGO2, as well as expressions of p-EGFR, p-Src and Bcl-2 in the brain. LINGO2 siRNA knockdown or EGFR inhibitor reversed the effects of delayed recanalization on apoptosis and brain expressions of LINGO2, p-EGFR, p-Src and Bcl-2 in rMCAO rats. EGFR activator abolished the deleterious effects of LINGO2 siRNA. In conclusion, our investigation demonstrated for the first time that delayed recanalization may enhance the entry of liver-derived TFF3 into ischemic brain upon restoring blood flow after MCAO, which attenuated neuronal apoptosis and neurological deficits at least in part via activating LINGO2/EGFR/Src pathway.
Collapse
Affiliation(s)
- Dujuan Li
- Department of Pathology, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University, People's Hospital of Henan University), Zhengzhou 450003, China; Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Lifei Lian
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Lei Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92354, USA
| | - Marcin Gamdzyk
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yi Huang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Desislava Doycheva
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Gaigai Li
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Shufeng Yu
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Yong Guo
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurosurgery, Henan Provincial People's Hospital, Zhengzhou 450003, China
| | - Ruiqing Kang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Hong Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Jiping Tang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA
| | - Lingfei Kong
- Department of Pathology, Henan Provincial People's Hospital (People's Hospital of Zhengzhou University, People's Hospital of Henan University), Zhengzhou 450003, China.
| | - John H Zhang
- Department of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA 92354, USA; Department of Neurosurgery, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
16
|
Zhang Y, Yang Y, Li H, Feng Q, Ge W, Xu X. Investigating the Potential Mechanisms and Therapeutic Targets of Inflammatory Cytokines in Post-stroke Depression. Mol Neurobiol 2024; 61:132-147. [PMID: 37592185 DOI: 10.1007/s12035-023-03563-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023]
Abstract
Post-stroke depression (PSD) affects approximately one-third of stroke survivors, severely impacting general recovery and quality of life. Despite extensive studies, the exact mechanisms underlying PSD remain elusive. However, emerging evidence implicates proinflammatory cytokines, including interleukin-1β, interleukin-6, tumor necrosis factor-alpha, and interleukin-18, play critical roles in PSD development. These cytokines contribute to PSD through various mechanisms, including hypothalamic-pituitary-adrenal (HPA) axis dysfunction, neurotransmitter alterations, neurotrophic factor changes, gut microbiota imbalances, and genetic predispositions. This review is aimed at exploring the role of cytokines in stroke and PSD while identifying their potential as specific therapeutic targets for managing PSD. A more profound understanding of the mechanisms regulating inflammatory cytokine expression and anti-inflammatory cytokines like interleukin-10 in PSD may facilitate the development of innovative interventions to improve outcomes for stroke survivors experiencing depression.
Collapse
Affiliation(s)
- Yutong Zhang
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Yuehua Yang
- Department of Neurology, Suzhou Yongding Hospital, Suzhou, 215028, China
| | - Hao Li
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Qian Feng
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China
| | - Wei Ge
- Department of Neurology, the Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221600, China.
| | - Xingshun Xu
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, 215000, China.
- Institute of Neuroscience, Soochow University, Suzhou, 215123, China.
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Soochow University, Suzhou, 215123, Jiangsu, China.
| |
Collapse
|
17
|
Furman M, Sihotsky V, Virag M, Kopolovets I, Nemethova M, Mucha R. Quantitative analysis of selected genetic markers of induced brain stroke ischemic tolerance detected in human blood. Brain Res 2023; 1821:148590. [PMID: 37739332 DOI: 10.1016/j.brainres.2023.148590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 09/24/2023]
Abstract
A brain stroke is a serious disease and the second leading cause of death in the European Union. Carotid stenosis accounts for 15% of all ischemic cerebral strokes. However, there is currently no effective screening for carotid disease. Analysis of the DNA from peripheral blood is increasingly being used for several disease diagnoses. The potentially beneficial therapeutic method of inducing tissue tolerance to ischemia has so far been studied mainly in animal models. The aim of this study is to investigate changes in the gene expression of selected markers of brain ischemia during carotid endarterectomy, considered in this study as an activator of ischemic tolerance. During the carotid endarterectomy, there is a short-term occlusion of the internal carotid artery. Using the RT-qPCR method, we detected changes in the early identified gene markers of brain ischemia (ADM, CDKN1A, GADD45G, IL6, TM4SF1) in peripheral blood during sub lethal cerebral ischemia caused by carotid endarterectomy. Patients underwenting surgical procedure were divided into three groups: asymptomatic, symptomatic, and those who underwent carotid endarterectomy after an acute stroke. The results were compared to a negative/control group. Carotid endarterectomy had an impact on the expression of all monitored biomarkers. We observed statistically significant changes (p value 0.05-0.001) when comparing the groups among themselves, as well as the presence of ischemic tolerance of brain tissue to ischemic attacks. In conclusion, ADM, GADD45G, and TM4SF1 were affected in symptomatic patients, GADD45G and IL6 in acute patients, and CDKN1A and ADM in asymptomatic group after application of carotid endarterectomy.
Collapse
Affiliation(s)
- Marek Furman
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Vladimir Sihotsky
- Eastern Slovak Institute of Cardiovascular Diseases and Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Ondavska 8, 040 01 Kosice, Slovakia
| | - Michal Virag
- Eastern Slovak Institute of Cardiovascular Diseases and Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Ondavska 8, 040 01 Kosice, Slovakia
| | - Ivan Kopolovets
- Eastern Slovak Institute of Cardiovascular Diseases and Faculty of Medicine, Pavol Jozef Safarik University, Kosice, Ondavska 8, 040 01 Kosice, Slovakia
| | - Miroslava Nemethova
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia
| | - Rastislav Mucha
- Institute of Neurobiology of Biomedical Research Center, Slovak Academy of Sciences, Soltesovej 4, 040 01 Kosice, Slovakia.
| |
Collapse
|
18
|
Berrino E, Carradori S, Carta F, Melfi F, Gallorini M, Poli G, Tuccinardi T, Fernández-Bolaños JG, López Ó, Petzer JP, Petzer A, Guglielmi P, Secci D, Supuran CT. A Multitarget Approach against Neuroinflammation: Alkyl Substituted Coumarins as Inhibitors of Enzymes Involved in Neurodegeneration. Antioxidants (Basel) 2023; 12:2044. [PMID: 38136164 PMCID: PMC10740956 DOI: 10.3390/antiox12122044] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/14/2023] [Accepted: 11/21/2023] [Indexed: 12/24/2023] Open
Abstract
Neurodegenerative disorders (NDs) include a large range of diseases characterized by neural dysfunction with a multifactorial etiology. The most common NDs are Alzheimer's disease and Parkinson's disease, in which cholinergic and dopaminergic systems are impaired, respectively. Despite different brain regions being affected, oxidative stress and inflammation were found to be common triggers in the pathogenesis and progression of both diseases. By taking advantage of a multi-target approach, in this work we explored alkyl substituted coumarins as neuroprotective agents, capable to reduce oxidative stress and inflammation by inhibiting enzymes involved in neurodegeneration, among which are Carbonic Anhydrases (CAs), Monoamine Oxidases (MAOs), and Cholinesterases (ChEs). The compounds were synthesized and profiled against the three targeted enzymes. The binding mode of the most promising compounds (7 and 9) within MAO-A and -B was analyzed through molecular modeling studies, providing and explanation for the different selectivities observed for the MAO isoforms. In vitro biological studies using LPS-stimulated rat astrocytes showed that some compounds were able to counteract the oxidative stress-induced neuroinflammation and hamper interleukin-6 secretion, confirming the success of this multitarget approach.
Collapse
Affiliation(s)
- Emanuela Berrino
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (E.B.); (P.G.); (D.S.)
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy; (F.C.); (C.T.S.)
| | - Simone Carradori
- Department of Pharmacy, ‘‘G. d’Annunzio” University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy; (F.M.); (M.G.)
| | - Fabrizio Carta
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy; (F.C.); (C.T.S.)
| | - Francesco Melfi
- Department of Pharmacy, ‘‘G. d’Annunzio” University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy; (F.M.); (M.G.)
| | - Marialucia Gallorini
- Department of Pharmacy, ‘‘G. d’Annunzio” University of Chieti-Pescara, via dei Vestini 31, 66100 Chieti, Italy; (F.M.); (M.G.)
| | - Giulio Poli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (G.P.); (T.T.)
| | - Tiziano Tuccinardi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; (G.P.); (T.T.)
| | - José G. Fernández-Bolaños
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, 41012 Seville, Spain; (J.G.F.-B.); (Ó.L.)
| | - Óscar López
- Departamento de Química Orgánica, Facultad de Química, Universidad de Sevilla, Apartado 1203, 41012 Seville, Spain; (J.G.F.-B.); (Ó.L.)
| | - Jacobus P. Petzer
- Pharmaceutical Chemistry, School of Pharmacy and Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.P.P.); (A.P.)
| | - Anél Petzer
- Pharmaceutical Chemistry, School of Pharmacy and Centre of Excellence for Pharmaceutical Sciences, North-West University, Potchefstroom 2531, South Africa; (J.P.P.); (A.P.)
| | - Paolo Guglielmi
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (E.B.); (P.G.); (D.S.)
| | - Daniela Secci
- Department of Drug Chemistry and Technologies, Sapienza University of Rome, P.le A. Moro 5, 00185 Rome, Italy; (E.B.); (P.G.); (D.S.)
| | - Claudiu T. Supuran
- NEUROFARBA Department, Sezione di Scienze Farmaceutiche e Nutraceutiche, Università degli Studi di Firenze, Via Ugo Schiff 6, 50019 Florence, Italy; (F.C.); (C.T.S.)
| |
Collapse
|
19
|
Telianidis J, Hunter A, Widdop R, Kemp-Harper B, Pham V, McCarthy C, Chai SY. Inhibition of insulin-regulated aminopeptidase confers neuroprotection in a conscious model of ischemic stroke. Sci Rep 2023; 13:19722. [PMID: 37957163 PMCID: PMC10643421 DOI: 10.1038/s41598-023-46072-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Stroke is a leading cause of mortality and morbidity with a paucity of effective pharmacological treatments. We have previously identified insulin-regulated aminopeptidase (IRAP) as a potential target for the development of a new class of drugs for the treatment of stroke, as global deletion of this gene in mice significantly protected against ischemic damage. In the current study, we demonstrate that small molecular weight IRAP inhibitors reduce infarct volume and improve neurological outcome in a hypertensive animal model of ischemic stroke. The effects of two structurally distinct IRAP inhibitors (HFI419 or SJM164) were investigated in a model of stroke where the middle cerebral artery was transiently occluded with endothelin-1 in the conscious spontaneously hypertensive rat. IRAP inhibitor was administered into the lateral ventricle at 2 or 6 h after stroke, with subsequent doses delivered at 24, 48 and 70 h post-stroke. Functional outcomes were assessed prior to drug treatment, and on day 1 and 3 post-stroke. Histological analyses and neuroinflammatory cytokine profiling were conducted at 72 and 24 h post-stroke respectively. IRAP inhibitor treatment following stroke significantly reduced infarct volume and improved neurological and motor deficits. These protective effects were maintained even when the therapeutic window was extended to 6 h. Examination of the cellular architecture at 72 h post-stroke demonstrated that IRAP expression was upregulated in CD11b positive cells and activated astrocytes. Furthermore, IRAP inhibitor treatment significantly increased gene expression for interleukin 6 and C-C motif chemokine ligand 2 in the ischemic core. This study provides proof-of-principle that selective inhibition of IRAP activity with two structurally distinct IRAP inhibitors reduces infarct volume and improves functional outcome even when the first dose is administered 6 h post-stroke. This is the first direct evidence that IRAP inhibitors are a class of drug with potential use in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Jonathon Telianidis
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Andrew Hunter
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Robert Widdop
- Department Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Barbara Kemp-Harper
- Department Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Vi Pham
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, VIC, 3052, Australia
| | - Claudia McCarthy
- Department Pharmacology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia
| | - Siew Yeen Chai
- Department of Physiology, Monash Biomedicine Discovery Institute, Monash University, Clayton, VIC, 3800, Australia.
| |
Collapse
|
20
|
Côrtes I, Alves G, Claudio-Da-Silva C, Baptista LS. Mimicking lipolytic, adipogenic, and secretory capacities of human subcutaneous adipose tissue by spheroids from distinct subpopulations of adipose stromal/stem cells. Front Cell Dev Biol 2023; 11:1219218. [PMID: 37842092 PMCID: PMC10570415 DOI: 10.3389/fcell.2023.1219218] [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: 05/08/2023] [Accepted: 09/11/2023] [Indexed: 10/17/2023] Open
Abstract
Background: Adipose tissue engineering may provide 3D models for the understanding of diseases such as obesity and type II diabetes. Recently, distinct adipose stem/stromal cell (ASC) subpopulations were identified from subcutaneous adipose tissue (SAT): superficial (sSAT), deep (dSAT), and the superficial retinacula cutis (sRC). This study aimed to test these subpopulations ASCs in 3D spheroid culture induced for adipogenesis under a pro-inflammatory stimulus with lipopolysaccharide (LPS). Methods: The samples of abdominal human subcutaneous adipose tissue were obtained during plastic aesthetic surgery (Protocol 145/09). Results: ASC spheroids showed high response to adipogenic induction in sSAT. All ASC spheroids increased their capacity to lipolysis under LPS. However, spheroids from dSAT were higher than from sSAT (p = 0.0045) and sRC (p = 0.0005). Newly formed spheroids and spheroids under LPS stimulus from sSAT showed the highest levels of fatty acid-binding protein 4 (FABP4) and CCAAT/enhancer-binding protein-α (C/EBPα) mRNA expression compared with dSAT and sRC (p < 0.0001). ASC spheroids from sRC showed the highest synthesis of angiogenic cytokines such as vascular endothelial growth factor (VEGF) compared with dSAT (p < 0.0228). Under LPS stimulus, ASC spheroids from sRC showed the highest synthesis of pro-inflammatory cytokines such as IL-6 compared with dSAT (p < 0.0092). Conclusion: Distinct physiological properties of SAT can be recapitulated in ASC spheroids. In summary, the ASC spheroid from dSAT showed the greatest lipolytic capacity, from sSAT the greatest adipogenic induction, and sRC showed greater secretory capacity when compared to the dSAT. Together, all these capacities form a true mimicry of SAT and hold the potential to contribute for a deeper understanding of cellular and molecular mechanisms in healthy and unhealthy adipose tissue scenarios or in response to pharmacological interventions.
Collapse
Affiliation(s)
- Isis Côrtes
- Federal University of Rio de Janeiro, Campus UFRJ Duque de Caxias Professor Geraldo Cidade, Rio de Janeiro, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
| | - Gutemberg Alves
- Cell and Molecular Biology Department, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - Cesar Claudio-Da-Silva
- Plastic Surgery Service, Clementino Fraga Filho University Hospital, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Leandra Santos Baptista
- Federal University of Rio de Janeiro, Campus UFRJ Duque de Caxias Professor Geraldo Cidade, Rio de Janeiro, Brazil
- Laboratory of Tissue Bioengineering, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
- Post-graduation Program in Biotechnology, National Institute of Metrology, Quality and Technology (Inmetro), Rio de Janeiro, Brazil
| |
Collapse
|
21
|
Li H, He C, Zhu R, Chen FM, Wang L, Leung FP, Tian XY, Tse G, Wong WT. Type 2 cytokines promote angiogenesis in ischemic muscle via endothelial IL-4Rα signaling. Cell Rep 2023; 42:112964. [PMID: 37556326 DOI: 10.1016/j.celrep.2023.112964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 05/21/2023] [Accepted: 07/26/2023] [Indexed: 08/11/2023] Open
Abstract
Peripheral arterial disease (PAD) is one of the leading causes of cardiovascular morbidity and mortality worldwide, yet current trials on therapeutic angiogenesis remain suboptimal. Type 2 immunity is critical for post-ischemic regeneration, but its regulatory role in revascularization is poorly characterized. Here, we show that type 2 cytokines, interleukin-4 (IL-4) and interleukin-13 (IL-13), are the key mediators in post-ischemic angiogenesis. IL-4/IL-13-deficient mice exhibit impaired reperfusion and muscle repair in an experimental model of PAD. We find that deletion of IL-4Rα in the endothelial compartment, rather than the myeloid compartment, leads to remarkable impairment in revascularization. Mechanistically, IL-4/IL-13 promote endothelial cell proliferation, migration, and tube formation via IL-4Rα/STAT6 signaling. Furthermore, attenuated IL-4/IL-13 expression is associated with the angiogenesis deficit in the setting of diabetic PAD, while IL-4/IL-13 treatment rescues this defective regeneration. Our findings reveal the therapeutic potential of type 2 cytokines in treating patients with muscle ischemia.
Collapse
Affiliation(s)
- Huixian Li
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| | - Chufeng He
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Ruiwen Zhu
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Francis M Chen
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Lin Wang
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Fung Ping Leung
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Gary Tse
- School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong 999077, China; Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, China
| | - Wing Tak Wong
- School of Life Sciences, Faculty of Science, The Chinese University of Hong Kong, Hong Kong 999077, China; State Key Laboratory of Agrobiotechnology, The Chinese University of Hong Kong, Hong Kong 999077, China.
| |
Collapse
|
22
|
Choi BR, Johnson KR, Maric D, McGavern DB. Monocyte-derived IL-6 programs microglia to rebuild damaged brain vasculature. Nat Immunol 2023; 24:1110-1123. [PMID: 37248420 PMCID: PMC11531796 DOI: 10.1038/s41590-023-01521-1] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 04/25/2023] [Indexed: 05/31/2023]
Abstract
Cerebrovascular injury (CVI) is a common pathology caused by infections, injury, stroke, neurodegeneration and autoimmune disease. Rapid resolution of a CVI requires a coordinated innate immune response. In the present study, we sought mechanistic insights into how central nervous system-infiltrating monocytes program resident microglia to mediate angiogenesis and cerebrovascular repair after an intracerebral hemorrhage. In the penumbrae of human stroke brain lesions, we identified a subpopulation of microglia that express vascular endothelial growth factor A. These cells, termed 'repair-associated microglia' (RAMs), were also observed in a rodent model of CVI and coexpressed interleukin (IL)-6Ra. Cerebrovascular repair did not occur in IL-6 knockouts or in mice lacking microglial IL-6Ra expression and single-cell transcriptomic analyses revealed faulty RAM programming in the absence of IL-6 signaling. Infiltrating CCR2+ monocytes were the primary source of IL-6 after a CVI and were required to endow microglia with proliferative and proangiogenic properties. Faulty RAM programming in the absence of IL-6 or inflammatory monocytes resulted in poor cerebrovascular repair, neuronal destruction and sustained neurological deficits that were all restored via exogenous IL-6 administration. These data provide a molecular and cellular basis for how monocytes instruct microglia to repair damaged brain vasculature and promote functional recovery after injury.
Collapse
Affiliation(s)
- Bo-Ran Choi
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Kory R Johnson
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dragan Maric
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA
| | - Dorian B McGavern
- Viral Immunology and Intravital Imaging Section, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
23
|
Abstract
Owing to its high disability and mortality rates, stroke has been the second leading cause of death worldwide. Since the pathological mechanisms of stroke are not fully understood, there are few clinical treatment strategies available with an exception of tissue plasminogen activator (tPA), the only FDA-approved drug for the treatment of ischemic stroke. Angiogenesis is an important protective mechanism that promotes neural regeneration and functional recovery during the pathophysiological process of stroke. Thus, inducing angiogenesis in the peri-infarct area could effectively improve hemodynamics, and promote vascular remodeling and recovery of neurovascular function after ischemic stroke. In this review, we summarize the cellular and molecular mechanisms affecting angiogenesis after cerebral ischemia registered in PubMed, and provide pro-angiogenic strategies for exploring the treatment of ischemic stroke, including endothelial progenitor cells, mesenchymal stem cells, growth factors, cytokines, non-coding RNAs, etc.
Collapse
Affiliation(s)
- Jie Fang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Zhi Wang
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University / Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
24
|
Dorschel KB, Wanebo JE. Physiological and pathophysiological mechanisms of the molecular and cellular biology of angiogenesis and inflammation in moyamoya angiopathy and related vascular diseases. Front Neurol 2023; 14:661611. [PMID: 37273690 PMCID: PMC10236939 DOI: 10.3389/fneur.2023.661611] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 01/16/2023] [Indexed: 06/06/2023] Open
Abstract
Rationale The etiology and pathophysiological mechanisms of moyamoya angiopathy (MMA) remain largely unknown. MMA is a progressive, occlusive cerebrovascular disorder characterized by recurrent ischemic and hemorrhagic strokes; with compensatory formation of an abnormal network of perforating blood vessels that creates a collateral circulation; and by aberrant angiogenesis at the base of the brain. Imbalance of angiogenic and vasculogenic mechanisms has been proposed as a potential cause of MMA. Moyamoya vessels suggest that aberrant angiogenic, arteriogenic, and vasculogenic processes may be involved in the pathophysiology of MMA. Circulating endothelial progenitor cells have been hypothesized to contribute to vascular remodeling in MMA. MMA is associated with increased expression of angiogenic factors and proinflammatory molecules. Systemic inflammation may be related to MMA pathogenesis. Objective This literature review describes the molecular mechanisms associated with cerebrovascular dysfunction, aberrant angiogenesis, and inflammation in MMA and related cerebrovascular diseases along with treatment strategies and future research perspectives. Methods and results References were identified through a systematic computerized search of the medical literature from January 1, 1983, through July 29, 2022, using the PubMed, EMBASE, BIOSIS Previews, CNKI, ISI web of science, and Medline databases and various combinations of the keywords "moyamoya," "angiogenesis," "anastomotic network," "molecular mechanism," "physiology," "pathophysiology," "pathogenesis," "biomarker," "genetics," "signaling pathway," "blood-brain barrier," "endothelial progenitor cells," "endothelial function," "inflammation," "intracranial hemorrhage," and "stroke." Relevant articles and supplemental basic science articles almost exclusively published in English were included. Review of the reference lists of relevant publications for additional sources resulted in 350 publications which met the study inclusion criteria. Detection of growth factors, chemokines, and cytokines in MMA patients suggests the hypothesis of aberrant angiogenesis being involved in MMA pathogenesis. It remains to be ascertained whether these findings are consequences of MMA or are etiological factors of MMA. Conclusions MMA is a heterogeneous disorder, comprising various genotypes and phenotypes, with a complex pathophysiology. Additional research may advance our understanding of the pathophysiology involved in aberrant angiogenesis, arterial stenosis, and the formation of moyamoya collaterals and anastomotic networks. Future research will benefit from researching molecular pathophysiologic mechanisms and the correlation of clinical and basic research results.
Collapse
Affiliation(s)
- Kirsten B. Dorschel
- Medical Faculty, Heidelberg University Medical School, Ruprecht-Karls-Universität Heidelberg, Heidelberg, Germany
| | - John E. Wanebo
- Department of Neurosurgery, Barrow Neurological Institute, St. Joseph's Hospital and Medical Center, Phoenix, AZ, United States
- Department of Neuroscience, HonorHealth Research Institute, Scottsdale, AZ, United States
| |
Collapse
|
25
|
Tirandi A, Sgura C, Carbone F, Montecucco F, Liberale L. Inflammatory biomarkers of ischemic stroke. Intern Emerg Med 2023; 18:723-732. [PMID: 36745280 PMCID: PMC10082112 DOI: 10.1007/s11739-023-03201-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 01/09/2023] [Indexed: 02/07/2023]
Abstract
Ischemic stroke remains the second leading cause of death and among the major causes of morbidity worldwide. Therapeutic options are currently limited to early reperfusion strategies, while pharmacological neuroprotective strategies despite showing promising results in the experimental setting constantly failed to enter the clinical arena. Inflammation plays an important role in the pathophysiology of ischemic stroke and mediators of inflammation have been longtime investigated as possible prognostic marker and therapeutic target for stroke patients. Here, we summarized available evidence on the role of cytokines, soluble adhesion molecules and adipokines in the pathophysiology, prognosis and therapy of ischemic stroke.
Collapse
Affiliation(s)
- Amedeo Tirandi
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Cosimo Sgura
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
| | - Federico Carbone
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| | - Fabrizio Montecucco
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy.
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Luca Liberale
- Department of Internal Medicine, University of Genoa, Viale Benedetto XV, 6, 16132, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa, Italian Cardiovascular Network, Largo Rosanna Benzi 10, 16132, Genoa, Italy
| |
Collapse
|
26
|
Shichita T, Ooboshi H, Yoshimura A. Neuroimmune mechanisms and therapies mediating post-ischaemic brain injury and repair. Nat Rev Neurosci 2023; 24:299-312. [PMID: 36973481 DOI: 10.1038/s41583-023-00690-0] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2023] [Indexed: 03/29/2023]
Abstract
The nervous and immune systems control whole-body homeostasis and respond to various types of tissue injury, including stroke, in a coordinated manner. Cerebral ischaemia and subsequent neuronal cell death activate resident or infiltrating immune cells, which trigger neuroinflammation that affects functional prognosis after stroke. Inflammatory immune cells exacerbate ischaemic neuronal injury after the onset of brain ischaemia; however, some of the immune cells thereafter change their function to neural repair. The recovery processes after ischaemic brain injury require additional and close interactions between the nervous and immune systems through various mechanisms. Thus, the brain controls its own inflammation and repair processes after injury via the immune system, which provides a promising therapeutic opportunity for stroke recovery.
Collapse
Affiliation(s)
- Takashi Shichita
- Stroke Renaissance Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan.
- Department of Neuroinflammation and Repair, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan.
- Core Research for Evolutionary Medical Science and Technology (CREST), Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.
| | - Hiroaki Ooboshi
- Section of Internal Medicine, Department of Medicine, Fukuoka Dental College, Fukuoka, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Shinjuku-ku, Tokyo, Japan
| |
Collapse
|
27
|
Yan H, Kawano T, Kanki H, Nishiyama K, Shimamura M, Mochizuki H, Sasaki T. Role of Polymorphonuclear Myeloid-Derived Suppressor Cells and Neutrophils in Ischemic Stroke. J Am Heart Assoc 2023; 12:e028125. [PMID: 36892072 PMCID: PMC10111556 DOI: 10.1161/jaha.122.028125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Background Immune cells play a vital role in the pathology of ischemic stroke. Neutrophils and polymorphonuclear myeloid-derived suppressor cells share a similar phenotype and have attracted increasing attention in immune regulation research, yet their dynamics in ischemic stroke remain elusive. Methods and Results Mice were randomly divided into 2 groups and intraperitoneally treated with anti-Ly6G (lymphocyte antigen 6 complex locus G) monoclonal antibody or saline. Distal middle cerebral artery occlusion and transient middle cerebral artery occlusion were applied to induce experimental stroke, and mice mortality was recorded until 28 days after stroke. Green fluorescent nissl staining was used to measure infarct volume. Cylinder and foot fault tests were used to evaluate neurological deficits. Immunofluorescence staining was conducted to confirm Ly6G neutralization and detect activated neutrophils and CD11b+Ly6G+ cells. Fluorescence-activated cell sorting was performed to evaluate polymorphonuclear myeloid-derived suppressor cell accumulation in brains and spleens after stroke. Anti-Ly6G antibody successfully depleted Ly6G expression in mice cortex but did not alter cortical physiological vasculature. Prophylactic anti-Ly6G antibody treatment ameliorated ischemic stroke outcomes in the subacute phase. Moreover, using immunofluorescence staining, we found that anti-Ly6G antibody suppressed activated neutrophil infiltration into parenchyma and decreased neutrophil extracellular trap formation in penumbra after stroke. Additionally, prophylactic anti-Ly6G antibody treatment reduced polymorphonuclear myeloid-derived suppressor cell accumulation in the ischemic hemisphere. Conclusions Our study suggested a protective effect of prophylactic anti-Ly6G antibody administration against ischemic stroke by reducing activated neutrophil infiltration and neutrophil extracellular trap formation in parenchyma and suppressing polymorphonuclear myeloid-derived suppressor cell accumulation in the brain. This study may provide a novel therapeutic approach for ischemic stroke.
Collapse
Affiliation(s)
- Haomin Yan
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
| | - Tomohiro Kawano
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
| | - Hideaki Kanki
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
| | - Kumiko Nishiyama
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
| | - Munehisa Shimamura
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
- Department of Health Development and Medicine Osaka University Graduate School of Medicine Osaka Japan
| | - Hideki Mochizuki
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
| | - Tsutomu Sasaki
- Department of Neurology, Graduate School of Medicine Osaka University Osaka Japan
- Department of Neurotherapeutics, Graduate School of Medicine Osaka University Osaka Japan
| |
Collapse
|
28
|
Carlsson R, Enström A, Paul G. Molecular Regulation of the Response of Brain Pericytes to Hypoxia. Int J Mol Sci 2023; 24:5671. [PMID: 36982744 PMCID: PMC10053233 DOI: 10.3390/ijms24065671] [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: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/14/2023] [Indexed: 03/18/2023] Open
Abstract
The brain needs sufficient oxygen in order to function normally. This is achieved by a large vascular capillary network ensuring that oxygen supply meets the changing demand of the brain tissue, especially in situations of hypoxia. Brain capillaries are formed by endothelial cells and perivascular pericytes, whereby pericytes in the brain have a particularly high 1:1 ratio to endothelial cells. Pericytes not only have a key location at the blood/brain interface, they also have multiple functions, for example, they maintain blood-brain barrier integrity, play an important role in angiogenesis and have large secretory abilities. This review is specifically focused on both the cellular and the molecular responses of brain pericytes to hypoxia. We discuss the immediate early molecular responses in pericytes, highlighting four transcription factors involved in regulating the majority of transcripts that change between hypoxic and normoxic pericytes and their potential functions. Whilst many hypoxic responses are controlled by hypoxia-inducible factors (HIF), we specifically focus on the role and functional implications of the regulator of G-protein signaling 5 (RGS5) in pericytes, a hypoxia-sensing protein that is regulated independently of HIF. Finally, we describe potential molecular targets of RGS5 in pericytes. These molecular events together contribute to the pericyte response to hypoxia, regulating survival, metabolism, inflammation and induction of angiogenesis.
Collapse
Affiliation(s)
- Robert Carlsson
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Andreas Enström
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
| | - Gesine Paul
- Translational Neurology Group, Department of Clinical Science, Wallenberg Neuroscience Centre and Wallenberg Centre for Molecular Medicine, Lund University, 22184 Lund, Sweden
- Department of Neurology, Scania University Hospital, 22185 Lund, Sweden
| |
Collapse
|
29
|
Yu W, Gong E, Liu B, Zhou L, Che C, Hu S, Zhang Z, Liu J, Shi J. Hydrogel-mediated drug delivery for treating stroke. CHINESE CHEM LETT 2023. [DOI: 10.1016/j.cclet.2023.108205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
|
30
|
Chen X, Liu L, Zhong Y, Liu Y. Enriched environment promotes post-stroke angiogenesis through astrocytic interleukin-17A. Front Behav Neurosci 2023; 17:1053877. [PMID: 36873773 PMCID: PMC9979086 DOI: 10.3389/fnbeh.2023.1053877] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 02/01/2023] [Indexed: 02/18/2023] Open
Abstract
Objective Our previous studies have revealed that the protective effect of an enriched environment (EE) may be linked with astrocyte proliferation and angiogenesis. However, the relationship between astrocytes and angiogenesis under EE conditions still requires further study. The current research examined the neuroprotective effects of EE on angiogenesis in an astrocytic interleukin-17A (IL-17A)-dependent manner following cerebral ischemia/reperfusion (I/R) injury. Methods A rat model of ischemic stroke based on middle cerebral artery occlusion (MCAO) for 120 min followed by reperfusion was established, after which rats were housed in either EE or standard conditions. A set of behavior tests were conducted, including the modified neurological severity scores (mNSS) and the rotarod test. The infarct volume was evaluated by means of 2,3,5-Triphenyl tetrazolium chloride (TTC) staining. To evaluate the levels of angiogenesis, the protein levels of CD34 were examined by means of immunofluorescence and western blotting, while the protein and mRNA levels of IL-17A, vascular endothelial growth factor (VEGF), and the angiogenesis-associated factors interleukin-6 (IL-6), JAK2, and STAT3 were detected by western blotting and real-time quantitative PCR (RT-qPCR). Results We found that EE promoted functional recovery, reduced infarct volume, and enhanced angiogenesis compared to rats in standard conditions. IL-17A expression in astrocytes was also increased in EE rats. EE treatment increased the levels of microvascular density (MVD) and promoted the expression of CD34, VEGF, IL-6, JAK2, and STAT3 in the penumbra, while the intracerebroventricular injection of the IL-17A-neutralizing antibody in EE rats attenuated EE-mediated functional recovery and angiogenesis. Conclusion Our findings revealed a possible neuroprotective mechanism of astrocytic IL-17A in EE-mediated angiogenesis and functional recovery after I/R injury, which might provide the theoretical basis for EE in clinical practise for stroke patients and open up new ideas for the research on the neural repair mechanism mediated by IL-17A in the recovery phase of stroke.
Collapse
Affiliation(s)
- Xiuping Chen
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Lingling Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yingjun Zhong
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Yang Liu
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| |
Collapse
|
31
|
Combination of EPC-EXs and NPC-EXs with miR-126 and miR-210 overexpression produces better therapeutic effects on ischemic stroke by protecting neurons through the Nox2/ROS and BDNF/TrkB pathways. Exp Neurol 2023; 359:114235. [PMID: 36174747 DOI: 10.1016/j.expneurol.2022.114235] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 09/17/2022] [Accepted: 09/22/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUNDS/AIMS Neural progenitor cells (NPCs) and endothelial progenitor cell (EPCs) exhibit synergistical effects on protecting endothelial cell functions. MiR-126 and miR-210 can protect cell activities by regulating brain-derived neurotrophic factor (BDNF) and reactive oxygen species (ROS) production. Exosomes (EXs) mediate the beneficial effects of stem cells via delivering microRNAs (miRs). Here, we investigated the combination effects of EXs from EPCs (EPC-EXs) and NPCs (NPC-EXs), and determined whether these EXs with miR-126 (EPC-EXsmiR-126) and miR-210 overexpression (NPC-EXsmiR-210) had better effects on hypoxia/reoxygenation (H/R)-injured neurons and ischemic stroke (IS). METHODS Cultured neurons were subjected to hypoxia for 6 h and then co-cultured with culture medium, NPC-EXs, EPC-EXs, NPC-EXs + EPC-EXs or NPC-EXsmiR-210 + EPC-EXsmiR-126 under normoxia for 24 h. Cell apoptosis, ROS production, neurite outgrowth and BDNF level were analyzed. Permanent middle cerebral artery occlusion (MCAO) was performed on C57BL/6 mice to build IS model. The mice were injected with PBS or various EXs via tail vein 2 h after MCAO operation. After 24 h, infarct volume and neurological deficits score (NDS), neuronal apoptosis, ROS production and spine density of dendrites, and brain BDNF level were analyzed. For mechanism study, NADPH oxidase 2(Nox2) and BDNF receptor tyrosine kinase receptor B (TrkB) were determined, and TrkB inhibitor k-252a was used in in vitro and in vivo study. RESULTS 1) The level of miR-210 or miR-126 was increased after NPC-EXs or EPC-EXs treatment respectively. 2) In H/R-injured neurons, NPC-EXs or EPC-EXs decreased cell apoptosis and ROS production and promoted neurite outgrowth, which were associated with the downregulation of Nox2 and the increase of BDNF and p-TrkB/TrkB level. 3) In MCAO mice, NPC-EXs or EPC-EXs decreased infarct volume and NDS, reduced neural apoptosis and ROS production, and promoted the spine density of dendrites. The levels of Nox2, BDNF and p-TrkB/TrkB in mouse brain tissues changed in similar patterns as seen in the in vitro study. 4) In both cell and mouse models, combination of NPC-EXs and EPC-EXs was more effective than NPC-EXs or EPC-EXs alone on all of these effects. 5) EPC-EXsmiR-126 + NPC-EXsmiR-210 had better effects compared to NPC-EXs + EPC-EXs, which were inhibited by k-252a. CONCLUSION EPC-EXsmiR-126 combined NPC-EXsmiR-210 further orchestrate the combinative protective effects of EPC-EXs and NPC-EXs on IS, possibly by protecting H/R-injured neurons through the Nox2/ ROS and BDNF/TrkB pathways.
Collapse
|
32
|
Advances in Antibody-Based Therapeutics for Cerebral Ischemia. Pharmaceutics 2022; 15:pharmaceutics15010145. [PMID: 36678774 PMCID: PMC9866586 DOI: 10.3390/pharmaceutics15010145] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/18/2022] [Accepted: 12/29/2022] [Indexed: 01/04/2023] Open
Abstract
Cerebral ischemia is an acute disorder characterized by an abrupt reduction in blood flow that results in immediate deprivation of both glucose and oxygen. The main types of cerebral ischemia are ischemic and hemorrhagic stroke. When a stroke occurs, several signaling pathways are activated, comprising necrosis, apoptosis, and autophagy as well as glial activation and white matter injury, which leads to neuronal cell death. Current treatments for strokes include challenging mechanical thrombectomy or tissue plasminogen activator, which increase the danger of cerebral bleeding, brain edema, and cerebral damage, limiting their usage in clinical settings. Monoclonal antibody therapy has proven to be effective and safe in the treatment of a variety of neurological disorders. In contrast, the evidence for stroke therapy is minimal. Recently, Clone MTS510 antibody targeting toll-like receptor-4 (TLR4) protein, ASC06-IgG1 antibody targeting acid sensing ion channel-1a (ASIC1a) protein, Anti-GluN1 antibodies targeting N-methyl-D-aspartate (NMDA) receptor associated calcium influx, GSK249320 antibody targeting myelin-associated glycoprotein (MAG), anti-High Mobility Group Box-1 antibody targeting high mobility group box-1 (HMGB1) are currently under clinical trials for cerebral ischemia treatment. In this article, we review the current antibody-based pharmaceuticals for neurological diseases, the use of antibody drugs in stroke, strategies to improve the efficacy of antibody therapeutics in cerebral ischemia, and the recent advancement of antibody drugs in clinical practice. Overall, we highlight the need of enhancing blood-brain barrier (BBB) penetration for the improvement of antibody-based therapeutics in the brain, which could greatly enhance the antibody medications for cerebral ischemia in clinical practice.
Collapse
|
33
|
Monsour M, Croci DM, Agazzi S, Borlongan CV. Contemplating IL-6, a double-edged sword cytokine: Which side to use for stroke pathology? CNS Neurosci Ther 2022; 29:493-497. [PMID: 36478506 PMCID: PMC9873516 DOI: 10.1111/cns.14041] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 11/02/2022] [Accepted: 11/09/2022] [Indexed: 12/12/2022] Open
Abstract
Interleukin (IL)-6 is a unique cytokine due to its dual signaling, with one pathway being pro-inflammatory (trans) and the other homeostatic (classical). Both of these pathways have been implicated in neuroinflammation following stroke, with initial inflammatory mechanisms being protective and later anti-inflammatory signaling promoting ischemic tissue recovery. IL-6 plays a major role in stroke pathology. However, given these distinctive IL-6 signaling consequences, IL-6 is a difficult cytokine to target for stroke therapies. Recent research suggests that the ratio between the pro-inflammatory binary IL6:sIL6R complex and the inactive ternary IL6:sIL6R:sgp130 complex may be a novel way to measure IL-6 signaling at different time points following ischemic injury. This ratio may approximate functional consequences on individualized stroke therapies, allowing clinicians to determine whether IL-6 agonists or antagonists should be used at specific time points.
Collapse
Affiliation(s)
- Molly Monsour
- University of South Florida Morsani College of MedicineTampaFloridaUSA
| | - Davide M. Croci
- Department of Neurosurgery and Brain RepairUniversity of South Florida, Morsani College of MedicineTampaFloridaUSA
| | - Siviero Agazzi
- Department of Neurosurgery and Brain RepairUniversity of South Florida, Morsani College of MedicineTampaFloridaUSA
| | - Cesar V. Borlongan
- Center of Excellence for Aging and Brain RepairUniversity of South Florida Morsani College of MedicineTampaFloridaUSA
| |
Collapse
|
34
|
Probing Interleukin-6 in Stroke Pathology and Neural Stem Cell Transplantation. Int J Mol Sci 2022; 23:ijms232415453. [PMID: 36555094 PMCID: PMC9779061 DOI: 10.3390/ijms232415453] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 11/30/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Stem cell transplantation is historically understood as a powerful preclinical therapeutic following stroke models. Current clinical strategies including clot busting/retrieval are limited by their time windows (tissue plasminogen activator: 3-4 h) and inevitable reperfusion injuries. However, 24+ h post-stroke, stem cells reduce infarction size, improve neurobehavioral performance, and reduce inflammatory agents including interleukins. Typically, interleukin-6 (IL-6) is regarded as proinflammatory, and thus, preclinical studies often discuss it as beneficial for neurological recuperation when stem cells reduce IL-6's expression. However, some studies have also demonstrated neurological benefit with upregulation of IL-6 or preconditioning of stem cells with IL-6. This review specifically focuses on stem cells and IL-6, and their occasionally disparate, occasionally synergistic roles in the setting of ischemic cerebrovascular insults.
Collapse
|
35
|
Gaire BP. Microglia as the Critical Regulators of Neuroprotection and Functional Recovery in Cerebral Ischemia. Cell Mol Neurobiol 2022; 42:2505-2525. [PMID: 34460037 PMCID: PMC11421653 DOI: 10.1007/s10571-021-01145-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 08/25/2021] [Indexed: 12/13/2022]
Abstract
Microglial activation is considered as the critical pathogenic event in diverse central nervous system disorders including cerebral ischemia. Proinflammatory responses of activated microglia have been well reported in the ischemic brain and neuroinflammatory responses of activated microglia have been believed to be the potential therapeutic strategy. However, despite having proinflammatory roles, microglia can have significant anti-inflammatory roles and they are associated with the production of growth factors which are responsible for neuroprotection and recovery after ischemic injury. Microglia can directly promote neuroprotection by preventing ischemic infarct expansion and promoting functional outcomes. Indirectly, microglia are involved in promoting anti-inflammatory responses, neurogenesis, and angiogenesis in the ischemic brain which are crucial pathophysiological events for ischemic recovery. In fact, anti-inflammatory cytokines and growth factors produced by microglia can promote neuroprotection and attenuate neurobehavioral deficits. In addition, microglia regulate phagocytosis, axonal regeneration, blood-brain barrier protection, white matter integrity, and synaptic remodeling, which are essential for ischemic recovery. Microglia can also regulate crosstalk with neurons and other cell types to promote neuroprotection and ischemic recovery. This review mainly focuses on the roles of microglia in neuroprotection and recovery following ischemic injury. Furthermore, this review also sheds the light on the therapeutic potential of microglia in stroke patients.
Collapse
Affiliation(s)
- Bhakta Prasad Gaire
- Department of Neurology and Anesthesiology, Shock Trauma and Anesthesiology Research Center, University of Maryland School of Medicine, Baltimore, MD, 21201, USA.
| |
Collapse
|
36
|
Takashima M, Nakamura K, Kiyohara T, Wakisaka Y, Hidaka M, Takaki H, Yamanaka K, Shibahara T, Wakisaka M, Ago T, Kitazono T. Low-dose sodium-glucose cotransporter 2 inhibitor ameliorates ischemic brain injury in mice through pericyte protection without glucose-lowering effects. Commun Biol 2022; 5:653. [PMID: 35780235 PMCID: PMC9250510 DOI: 10.1038/s42003-022-03605-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 06/20/2022] [Indexed: 12/11/2022] Open
Abstract
Antidiabetic sodium-glucose cotransporter 2 (SGLT2) inhibitors have attracted attention for their cardiorenal-protective properties beyond their glucose-lowering effect. However, their benefits in ischemic stroke remain controversial. Here we show the effects of luseogliflozin, a selective SGLT2 inhibitor, in acute ischemic stroke, using a permanent middle cerebral artery occlusion (pMCAO) model in non-diabetic mice. Pretreatment with low-dose luseogliflozin, which does not affect blood glucose levels, significantly attenuated infarct volume, blood-brain barrier disruption, and motor dysfunction after pMCAO. SGLT2 was expressed predominantly in brain pericytes and was upregulated in peri- and intra-infarct areas. Notably, luseogliflozin pretreatment reduced pericyte loss in ischemic areas. In cultured pericytes, luseogliflozin activated AMP-activated protein kinase α and increased mitochondrial transcription factor A expression and number of mitochondria, conferring resistance to oxygen-glucose deprivation. Collectively, pre-stroke inhibition of SGLT2 induces ischemic tolerance in brain pericytes independent of the glucose-lowering effect, contributing to the attenuation of ischemic brain injury. Pre-treatment of non-diabetic mice with the SGLT2 inhibitor, luseogliflozin, reduces brain damage and neurological dysfunction following middle cerebral artery occlusion by acquiring ischemic tolerance in pericytes.
Collapse
Affiliation(s)
- Masamitsu Takashima
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kuniyuki Nakamura
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Takuya Kiyohara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinobu Wakisaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masaoki Hidaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Hayato Takaki
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kei Yamanaka
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tomoya Shibahara
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Masanori Wakisaka
- Wakisaka Internal Medicine Clinic, 1-24-19 Fujisaki, Sawara-ku, Fukuoka, 814-0013, Japan
| | - Tetsuro Ago
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takanari Kitazono
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
37
|
Click chemistry extracellular vesicle/peptide/chemokine nanomissiles for treating central nervous systems injuries. Acta Pharm Sin B 2022; 13:2202-2218. [DOI: 10.1016/j.apsb.2022.06.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/02/2022] [Accepted: 06/03/2022] [Indexed: 12/19/2022] Open
|
38
|
Rutin protects hemorrhagic stroke development via supressing oxidative stress and inflammatory events in a zebrafish model. Eur J Pharmacol 2022; 925:174973. [DOI: 10.1016/j.ejphar.2022.174973] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 03/11/2022] [Accepted: 04/19/2022] [Indexed: 01/06/2023]
|
39
|
Circulating ADAMTS13 Levels Are Associated with an Increased Occurrence of Obstructive Sleep Apnea. DISEASE MARKERS 2022; 2022:1504137. [PMID: 35392493 PMCID: PMC8983172 DOI: 10.1155/2022/1504137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 02/25/2022] [Indexed: 11/17/2022]
Abstract
Background and Aims. Obstructive sleep apnea (OSA) is strongly associated with obesity, metabolic diseases, coronary artery disease (CAD), stroke, hypertension, and other disorders. This study assessed the relationship between circulating a disintegrin and metalloprotease with a thrombospondin type 1 motif, member 13 (ADAMTS13) levels and the presence of OSA. Materials and Methods. This cross-sectional study included a total of 223 patients. We used a powerful high-throughput multiplexed immunobead-based assay to detect circulating levels of ADAMTS13. The associations between circulating ADAMTS13 levels and OSA were evaluated by multivariate logistic regression analysis. Results. Circulating ADAMTS13 levels were significantly elevated in patients with OSA compared with controls (0.8 vs. 2.7 μg/mL, respectively,
). After adjusting for confounding factors, circulating ADAMTS13 levels were significantly independently associated with the presence of OSA (
, 95% confidence interval (CI) =4.11–24.13,
). Furthermore, circulating ADAMTS13 levels showed discriminatory accuracy in assessing the presence of OSA (area under the curve: 0.87, 95% CI 0.81–0.93,
). Conclusion. Circulating ADAMTS13 levels were significantly correlated with the presence of OSA. ADAMTS13 may therefore function as a novel biomarker for monitoring the development and progression of OSA.
Collapse
|
40
|
Dandekar MP, Yin X, Peng T, Devaraj S, Morales R, McPherson DD, Huang SL. Repetitive xenon treatment improves post-stroke sensorimotor and neuropsychiatric dysfunction. J Affect Disord 2022; 301:315-330. [PMID: 34990636 DOI: 10.1016/j.jad.2022.01.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/21/2021] [Accepted: 01/02/2022] [Indexed: 12/20/2022]
Abstract
Stroke is a life-changing event as stroke survivors experience changes in personality, emotions and mood. We investigated the effect of xenon gas encapsulated in liposomes on stroke-generated sensorimotor impairments, and anxiety- and depression-like phenotypes. Ischemic stroke was created by the intraluminal middle cerebral artery occlusion (MCAO) for 6 h followed by reperfusion in rats. Xenon-liposome (6 mg/kg, intravenous) treatment was given multiple times starting at 2 h post-ischemia through 6 h (5X), and once-daily for next 3 days. Rats underwent ischemic injury displayed sensorimotor deficits in the adhesive removal, vibrissae-evoked forelimb placement and rotarod tests. These animals also made lesser entries and spent less time on open arms of the elevated-plus maze and swam more in passive mode in the forced swimming test, indicating anxiety- and depression-like behaviors at 28- and 35-days post-injury, respectively. Repeated intravenous treatment with xenon-liposomes ameliorated these behavioral aberrations (p < 0.05). Gut microbiome analysis (16S ribosomal-RNA gene sequencing) showed a decrease in the Clostridium clusters XI, XIVa, XVIII and Lactobacillus bacterium, and increase of the Prevotella in the xenon-liposome group. No microbiota communities were majorly affected across the treatments. Moreover, xenon treatment group showed augmented plasma levels of IL-6 cytokines (∼5 fold) on day-35 post-ischemia, while no change was noticed in the IL-1β, IL-4, IL-10, IL-13 and MCP-1 levels. Our data highlights the safety, behavioral recovery and reversal of post-stroke brain injury following xenon-liposome treatment in an extended ischemic model. These results show the potential for this treatment strategy to be translated to patients with stroke.
Collapse
Affiliation(s)
- Manoj P Dandekar
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth) McGovern Medical School, Houston, TX, USA; presently Manoj P. Dandekar is affiliated with Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Xing Yin
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth) McGovern Medical School, Houston, TX, USA
| | - Tao Peng
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth) McGovern Medical School, Houston, TX, USA
| | - Sridevi Devaraj
- Department of Pathology & Immunology, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Rodrigo Morales
- Department of Neurology, The University of Texas Health Science Center at Houston (UTHealth) McGovern Medical School, Houston, TX, USA; CIBQA, Universidad Bernardo O'Higgins. Santiago, Chile
| | - David D McPherson
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth) McGovern Medical School, Houston, TX, USA; Center for Clinical and Translational Sciences at The University of Texas Health Science Center at Houston, TX, USA
| | - Shao-Ling Huang
- Department of Internal Medicine, Division of Cardiology, The University of Texas Health Science Center at Houston (UTHealth) McGovern Medical School, Houston, TX, USA.
| |
Collapse
|
41
|
Ma Y, Yang S, He Q, Zhang D, Chang J. The Role of Immune Cells in Post-Stroke Angiogenesis and Neuronal Remodeling: The Known and the Unknown. Front Immunol 2022; 12:784098. [PMID: 34975872 PMCID: PMC8716409 DOI: 10.3389/fimmu.2021.784098] [Citation(s) in RCA: 70] [Impact Index Per Article: 23.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 11/29/2021] [Indexed: 12/12/2022] Open
Abstract
Following a cerebral ischemic event, substantial alterations in both cellular and molecular activities occur due to ischemia-induced cerebral pathology. Mounting evidence indicates that the robust recruitment of immune cells plays a central role in the acute stage of stroke. Infiltrating peripheral immune cells and resident microglia mediate neuronal cell death and blood-brain barrier disruption by releasing inflammation-associated molecules. Nevertheless, profound immunological effects in the context of the subacute and chronic recovery phase of stroke have received little attention. Early attempts to curtail the infiltration of immune cells were effective in mitigating brain injury in experimental stroke studies but failed to exert beneficial effects in clinical trials. Neural tissue damage repair processes include angiogenesis, neurogenesis, and synaptic remodeling, etc. Post-stroke inflammatory cells can adopt divergent phenotypes that influence the aforementioned biological processes in both endothelial and neural stem cells by either alleviating acute inflammatory responses or secreting a variety of growth factors, which are substantially involved in the process of angiogenesis and neurogenesis. To better understand the multiple roles of immune cells in neural tissue repair processes post stroke, we review what is known and unknown regarding the role of immune cells in angiogenesis, neurogenesis, and neuronal remodeling. A comprehensive understanding of these inflammatory mechanisms may help identify potential targets for the development of novel immunoregulatory therapeutic strategies that ameliorate complications and improve functional rehabilitation after stroke.
Collapse
Affiliation(s)
- Yinzhong Ma
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Shilun Yang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qianyan He
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Dianhui Zhang
- Department of Neurology, The First Hospital of Jilin University, Changchun, China
| | - Junlei Chang
- Shenzhen Key Laboratory of Biomimetic Materials and Cellular Immunomodulation, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| |
Collapse
|
42
|
Abstract
PURPOSE OF REVIEW To highlight recent developments in studying mechanisms by which the apolipoprotein E4 (APOE4) allele affects the metabolism of brain lipids and predisposes the brain to inflammation and Alzheimer's disease (AD) dementia. RECENT FINDINGS APOE4 activates Ca2+ dependent phospholipase A2 (cPLA2) leading to changes in arachidonic acid (AA), eicosapentaenoic acid and docosahexaenoic acid signaling cascades in the brain. Among these changes, the increased conversion of AA to eicosanoids associates with sustained and unresolved chronic brain inflammation. The effects of APOE4 on the brain differ by age, disease stage, nutritional status and can be uncovered by brain imaging studies of brain fatty acid uptake. Reducing cPLA2 expression in the dementia brain presents a viable strategy that awaits to be tested. SUMMARY Fatty acid brain imaging techniques can clarify how changes to brain polyunsaturated fatty acid metabolism during the various phases of AD and guide the development of small molecules to mitigate brain inflammation.
Collapse
Affiliation(s)
| | - Brandon Ebright
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy
| | - Hussein N Yassine
- Department of Neurology and Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
43
|
Kuffner MTC, Koch SP, Kirchner M, Mueller S, Lips J, An J, Mertins P, Dirnagl U, Endres M, Boehm-Sturm P, Harms C, Hoffmann CJ. Paracrine Interleukin 6 Induces Cerebral Remodeling at Early Stages After Unilateral Common Carotid Artery Occlusion in Mice. Front Cardiovasc Med 2022; 8:805095. [PMID: 35155612 PMCID: PMC8830347 DOI: 10.3389/fcvm.2021.805095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 12/20/2021] [Indexed: 12/26/2022] Open
Abstract
Aims Carotid artery disease is frequent and can result in chronic modest hypoperfusion of the brain. If no transient ischemic attack or stroke occur, it is classified asymptomatic. In the long-term, though, it can lead to cognitive impairment. Fostering cerebral remodeling after carotid artery occlusion might be a new concept of treatment. Paracrine Interleukin 6 (IL-6) can induce such remodeling processes at early stages. However, it has neurodegenerative long-term effects. With this exploratory study, we investigated the effect of paracrine IL-6 on cerebral remodeling in early stages after asymptomatic carotid artery occlusion to identify new treatment targets. Methods and Results To mimic a human asymptomatic carotid artery disease, we used a mouse model of unilateral common carotid artery (CCA) occlusion. We developed a mouse model for inducible paracrine cerebral IL-6 expression (Cx30-Cre-ERT2;FLEX-IL6) and induced IL-6 2 days after CCA occlusion. We studied the effects of paracrine IL-6 after CCA occlusion on neuronal connectivity using diffusion tensor imaging and on local proteome regulations of the hypo-perfused striatum and contralateral motor cortex using mass spectrometry of laser capture micro-dissected tissues. Paracrine IL-6 induced cerebral remodeling leading to increased inter-hemispheric connectivity and changes in motor system connectivity. We identified changes in local protein abundance which might have adverse effects on functional outcome such as upregulation of Synuclein gamma (Sncg) or downregulation of Proline Dehydrogenase 1 (Prodh). However, we also identified changes in local protein abundance having potentially beneficial effects such as upregulation of Caprin1 or downregulation of GABA transporter 1 (Gat1). Conclusions Paracrine cerebral IL-6 at early stages induces changes in motor system connectivity and the proteome after asymptomatic CCA occlusion. Our results may help to distinguish unfavorable from beneficial IL-6 dependent protein regulations. Focusing on these targets might generate new treatments to improve long-term outcome in patients with carotid artery disease.
Collapse
Affiliation(s)
- Melanie T. C. Kuffner
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Stefan P. Koch
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Marieluise Kirchner
- Core Unit Proteomics, Berlin Institute of Health at Charité- Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Susanne Mueller
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Janet Lips
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Jeehye An
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Philipp Mertins
- Core Unit Proteomics, Berlin Institute of Health at Charité- Universitätsmedizin Berlin, Max Delbrück Center for Molecular Medicine, Berlin, Germany
| | - Ulrich Dirnagl
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Partner Site Berlin, Berlin, Germany
- Einstein Center for Neuroscience, Berlin, Germany
- QUEST Quality, Ethics, Open Science, Translation, Center for Transforming Biomedical Research, Berlin Institute of Health, Berlin, Germany
| | - Matthias Endres
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- NeuroCure Clinical Research Center, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Neurodegenerative Diseases (DZNE), Partner Site Berlin, Berlin, Germany
- Einstein Center for Neuroscience, Berlin, Germany
| | - Philipp Boehm-Sturm
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Christoph Harms
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
- Einstein Center for Neuroscience, Berlin, Germany
- Christoph Harms
| | - Christian J. Hoffmann
- Klinik und Hochschulambulanz für Neurologie mit Experimenteller Neurologie, Charité-Universitätsmedizin Berlin, Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
- Center for Stroke Research Berlin, Charité-Universitätsmedizin Berlin, Berlin, Germany
- *Correspondence: Christian J. Hoffmann
| |
Collapse
|
44
|
Modifications of gene expression detected in peripheral blood after brain ischemia treated with remote postconditioning. Mol Biol Rep 2021; 49:477-485. [PMID: 34766231 DOI: 10.1007/s11033-021-06899-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/29/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND A stroke is an acute damage to a certain area of a nerve tissue of the brain. In developed countries, it ranks second among the most often causes of death and is also the leading cause of disability. Recent findings emphasize the significant neuroprotective effect of conditioning on the course and rate of recovery after ischemic attack; however the molecular mechanism of ischemic tolerance induced by conditioning is still not completely explored. METHODS AND RESULTS The purpose of this study is an identification of changes in gene expression induced by stimulation of reaction cascades after activation of the neuroprotective mechanism using an experimental rat model of global ischemia. The induction of neuroprotective cascades was stimulated by the application of early and delayed form of remote ischemic postconditioning. The quantitative qRT-PCR method was used to assess the rate of change in ADM, BDNF, CDKN1A, CREB, GADD45G, IL6, nNOS, and TM4SF1 gene expression levels 72 h after ischemic attack. The detected results confirm the neuroprotective effect of both forms of postconditioning. Participation of neuroprotection-related gene expression changes was observed once as an early one (CREB, GADD45G), once as a delayed one (ADM, IL6), or both (BDNF, CDKN1A, nNOS, TM4SF1) postconditioning forms, depending on the particular gene. CONCLUSIONS Our results characterize impact of ischemic tolerance on the molecular level. We predict ischemic tolerance to be consisted of complex combination of early and delayed remote postconditioning.
Collapse
|
45
|
Liberale L, Bonetti NR, Puspitasari YM, Vukolic A, Akhmedov A, Diaz‐Cañestro C, Keller S, Montecucco F, Merlini M, Semerano A, Giacalone G, Bacigaluppi M, Sessa M, Ruschitzka F, Lüscher TF, Libby P, Beer JH, Camici GG. TNF-α antagonism rescues the effect of ageing on stroke: Perspectives for targeting inflamm-ageing. Eur J Clin Invest 2021; 51:e13600. [PMID: 34076259 PMCID: PMC8596431 DOI: 10.1111/eci.13600] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/10/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
AIMS Epidemiologic evidence links ischemic stroke to age, yet the mechanisms that underlie the specific and independent effects of age on stroke remain elusive, impeding the development of targeted treatments. This study tested the hypothesis that age directly aggravates stroke outcomes and proposes inflamm-aging as a mediator and potential therapeutic target. METHODS 3 months- (young) and 18-20 months-old (old) mice underwent transient middle cerebral artery occlusion (tMCAO) for 30 minutes followed by 48 hours of reperfusion. Old animals received weekly treatment with the TNF-α neutralizing antibody adalimumab over 4 weeks before tMCAO in a separate set of experiments. Plasma levels of TNF- α were assessed in patients with ischemic stroke and correlated with age and outcome. RESULTS Old mice displayed larger stroke size than young ones with increased neuromotor deficit. Immunohistochemical analysis revealed impairment of the blood-brain barrier in old mice, i.e. increased post-stroke degradation of endothelial tight junctions and expression of tight junctions-digesting and neurotoxic matrix metalloproteinases. At baseline, old animals showed a broad modulation of several circulating inflammatory mediators. TNF-α displayed the highest increase in old animals and its inhibition restored the volume of stroke, neuromotor performance, and survival rates of old mice to the levels observed in young ones. Patients with ischemic stroke showed increased TNF-α plasma levels which correlated with worsened short-term neurological outcome as well as with age. CONCLUSIONS This study identifies TNF-α as a causative contributor to the deleterious effect of aging on stroke and points to inflamm-aging as a mechanism of age-related worsening of stroke outcomes and potential therapeutic target in this context. Thus, this work provides a basis for tailoring novel stroke therapies for the particularly vulnerable elderly population.
Collapse
Affiliation(s)
- Luca Liberale
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
- Department of Internal MedicineFirst Clinic of Internal MedicineUniversity of GenoaGenoaItaly
| | - Nicole R. Bonetti
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital of BadenBadenSwitzerland
| | | | - Ana Vukolic
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
| | | | | | - Stephan Keller
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
| | - Fabrizio Montecucco
- Department of Internal MedicineFirst Clinic of Internal MedicineUniversity of GenoaGenoaItaly
- IRCCS Ospedale Policlinico San Martino Genoa – Italian Cardiovascular NetworkGenoaItaly
| | - Mario Merlini
- Blood & Brain @ Caen–Normandie InstituteGIP CyceronCaenFrance
| | - Aurora Semerano
- Department of NeurologySan Raffaele Scientific InstituteMilanoItaly
| | | | | | - Maria Sessa
- Department of NeurologyPapa Giovanni XXIII HospitalBergamoItaly
| | - Frank Ruschitzka
- Department of CardiologyUniversity Heart CenterUniversity Hospital ZurichZurichSwitzerland
| | - Thomas F. Lüscher
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
- Royal Brompton and Harefield Hospitals and Imperial CollegeLondonUK
| | - Peter Libby
- Division of Cardiovascular MedicineDepartment of MedicineBrigham and Women’s HospitalHarvard Medical SchoolBostonMAUSA
| | - Jürg H. Beer
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
- Department of Internal MedicineCantonal Hospital of BadenBadenSwitzerland
| | - Giovanni G. Camici
- Center for Molecular CardiologyUniversity of ZürichSchlierenSwitzerland
- Department of CardiologyUniversity Heart CenterUniversity Hospital ZurichZurichSwitzerland
- Department of Research and EducationUniversity Hospital ZurichZurichSwitzerland
| |
Collapse
|
46
|
Kirzinger B, Stroux A, Rackoll T, Endres M, Flöel A, Ebinger M, Nave AH. Elevated Serum Inflammatory Markers in Subacute Stroke Are Associated With Clinical Outcome but Not Modified by Aerobic Fitness Training: Results of the Randomized Controlled PHYS-STROKE Trial. Front Neurol 2021; 12:713018. [PMID: 34512526 PMCID: PMC8426903 DOI: 10.3389/fneur.2021.713018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/28/2021] [Indexed: 12/02/2022] Open
Abstract
Background: Inflammatory markers, such as C-reactive Protein (CRP), Interleukin-6 (IL-6), tumor necrosis factor (TNF)-alpha and fibrinogen, are upregulated following acute stroke. Studies have shown associations of these biomarkers with increased mortality, recurrent vascular risk, and poor functional outcome. It is suggested that physical fitness training may play a role in decreasing long-term inflammatory activity and supports tissue recovery. Aim: We investigated the dynamics of selected inflammatory markers in the subacute phase following stroke and determined if fluctuations are associated with functional recovery up to 6 months. Further, we examined whether exposure to aerobic physical fitness training in the subacute phase influenced serum inflammatory markers over time. Methods: This is an exploratory analysis of patients enrolled in the multicenter randomized-controlled PHYS-STROKE trial. Patients within 45 days of stroke onset were randomized to receive either four weeks of aerobic physical fitness training or relaxation sessions. Generalized estimating equation models were used to investigate the dynamics of inflammatory markers and the associations of exposure to fitness training with serum inflammatory markers over time. Multiple logistic regression models were used to explore associations between inflammatory marker levels at baseline and three months after stroke and outcome at 3- or 6-months. Results: Irrespective of the intervention group, high sensitive CRP (hs-CRP), IL-6, and fibrinogen (but not TNF-alpha) were significantly lower at follow-up visits when compared to baseline (p all ≤ 0.01). In our cohort, exposure to aerobic physical fitness training did not influence levels of inflammatory markers over time. In multivariate logistic regression analyses, increased baseline IL-6 and fibrinogen levels were inversely associated with worse outcome at 3 and 6 months. Increased levels of hs-CRP at 3 months after stroke were associated with impaired outcome at 6 months. We found no independent associations of TNF-alpha levels with investigated outcome parameters. Conclusion: Serum markers of inflammation were elevated after stroke and decreased within 6 months. In our cohort, exposure to aerobic physical fitness training did not modify the dynamics of inflammatory markers over time. Elevated IL-6 and fibrinogen levels in early subacute stroke were associated with worse outcome up to 6-months after stroke. Clinical Trial Registration:ClinicalTrials.gov, NCT01953549.
Collapse
Affiliation(s)
- Bernadette Kirzinger
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Andrea Stroux
- Institute of Biometry and Clinical Epidemiology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Torsten Rackoll
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Berlin Institute of Health QUEST Center for Transforming Biomedical Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Matthias Endres
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,NeuroCure Clinical Research Center, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Klinik Und Hochschulambulanz für Neurologie, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,German Center for Neurodegenerative Diseases, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Agnes Flöel
- Department of Neurology, University Medicine Greifswald, Greifswald, Germany.,German Center for Neurodegenerative Diseases, Partner Site Rostock/Greifswald, Greifswald, Germany
| | - Martin Ebinger
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Medical Park Berlin Humboldtmühle, Berlin, Germany
| | - Alexander Heinrich Nave
- Center for Stroke Research Berlin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Klinik Und Hochschulambulanz für Neurologie, Charité - Universitätsmedizin Berlin, Berlin, Germany.,German Center for Cardiovascular Research, Partner Site Berlin, Berlin, Germany.,Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
47
|
Grossi C, Artusi C, Meroni P, Borghi MO, Neglia L, Lonati PA, Oggioni M, Tedesco F, De Simoni MG, Fumagalli S. β2 glycoprotein I participates in phagocytosis of apoptotic neurons and in vascular injury in experimental brain stroke. J Cereb Blood Flow Metab 2021; 41:2038-2053. [PMID: 33444093 PMCID: PMC8323337 DOI: 10.1177/0271678x20984551] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Beta-2 Glycoprotein I (β2-GPI) is the main target of anti-phospholipid antibodies (aPL) in the autoimmune anti-phospholipid syndrome, characterized by increased risk of stroke. We here investigated the antibody independent role of β2-GPI after ischemia/reperfusion, modeled in vivo by transient middle cerebral artery occlusion (tMCAo) in male C57Bl/6J mice; in vitro by subjecting immortalized human brain microvascular endothelial cells (ihBMEC) to 16 h hypoxia and 4 h re-oxygenation. ApoH (coding for β2-GPI) was upregulated selectively in the liver at 48 h after tMCAo. At the same time β2-GPI circulating levels increased. β2-GPI was detectable in brain parenchyma and endothelium at all time points after tMCAo. Parenchymal β2-GPI recognized apoptotic neurons (positive for annexin V, C3 and TUNEL) cleared by CD68+ brain macrophages. Hypoxic ihBMEC showed increased release of IL-6, over-expression of thrombomodulin and IL-1α after re-oxygenation with β2-GPI alone. β2-GPI interacted with mannose-binding lectin in mouse plasma and ihBMEC medium, potentially involved in formation of thrombi. We show for the first time that brain ischemia triggers the hepatic production of β2-GPI. β2-GPI is present in the ischemic endothelium, enhancing vascular inflammation, and extravasates binding stressed neurons before their clearance by phagocytosis. Thus β2-GPI may be a new mediator of brain injury following ischemic stroke.
Collapse
Affiliation(s)
- Claudia Grossi
- Istituto Auxologico Italiano, IRCCS, Laboratory of Immuno-Rheumatology, Milan, Italy
| | - Carolina Artusi
- Rheumatology Department, ASST Gaetano Pini-CTO, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - PierLuigi Meroni
- Istituto Auxologico Italiano, IRCCS, Laboratory of Immuno-Rheumatology, Milan, Italy
| | - Maria Orietta Borghi
- Istituto Auxologico Italiano, IRCCS, Laboratory of Immuno-Rheumatology, Milan, Italy.,Department of Clinical Sciences and Community Health, University of Milan, Milan, Italy
| | - Laura Neglia
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| | - Paola Adele Lonati
- Istituto Auxologico Italiano, IRCCS, Laboratory of Immuno-Rheumatology, Milan, Italy
| | - Marco Oggioni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| | - Francesco Tedesco
- Istituto Auxologico Italiano, IRCCS, Laboratory of Immuno-Rheumatology, Milan, Italy
| | - Maria-Grazia De Simoni
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| | - Stefano Fumagalli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Department of Neuroscience, Milan, Italy
| |
Collapse
|
48
|
Yoon Y, Voloudakis G, Doran N, Zhang E, Dimovasili C, Chen L, Shao Z, Darmanis S, Tang C, Tang J, Wang VX, Hof PR, Robakis NK, Georgakopoulos A. PS1 FAD mutants decrease ephrinB2-regulated angiogenic functions, ischemia-induced brain neovascularization and neuronal survival. Mol Psychiatry 2021; 26:1996-2012. [PMID: 32541930 PMCID: PMC7736163 DOI: 10.1038/s41380-020-0812-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/29/2020] [Accepted: 06/04/2020] [Indexed: 12/12/2022]
Abstract
Microvascular pathology and ischemic lesions contribute substantially to neuronal dysfunction and loss that lead to Alzheimer disease (AD). To facilitate recovery, the brain stimulates neovascularization of damaged tissue via sprouting angiogenesis, a process regulated by endothelial cell (EC) sprouting and the EphB4/ephrinB2 system. Here, we show that in cultures of brain ECs, EphB4 stimulates the VE-cadherin/Rok-α angiogenic complexes known to mediate sprouting angiogenesis. Importantly, brain EC cultures expressing PS1 FAD mutants decrease the EphB4-stimulated γ-secretase cleavage of ephrinB2 and reduce production of the angiogenic peptide ephrinB2/CTF2, the VE-cadherin angiogenic complexes and EC sprouting and tube formation. These data suggest that FAD mutants may attenuate ischemia-induced brain angiogenesis. Supporting this hypothesis, ischemia-induced VE-cadherin angiogenic complexes, levels of neoangiogenesis marker Endoglin, vascular density, and cerebral blood flow recovery, are all decreased in brains of mouse models expressing PS1 FAD mutants. Ischemia-induced brain neuronal death and cognitive deficits also increase in these mice. Furthermore, a small peptide comprising the C-terminal sequence of peptide ephrinB2/CTF2 rescues angiogenic functions of brain ECs expressing PS1 FAD mutants. Together, our data show that PS1 FAD mutations impede the EphB4/ephrinB2-mediated angiogenic functions of ECs and impair brain neovascularization, neuronal survival and cognitive recovery following ischemia. Furthermore, our data reveal a novel brain angiogenic mechanism targeted by PS1 FAD mutants and a potential therapeutic target for ischemia-induced neurodegeneration. Importantly, FAD mutant effects occur in absence of neuropathological hallmarks of AD, supporting that such hallmarks may form downstream of mutant effects on neoangiogenesis and neuronal survival.
Collapse
Affiliation(s)
- YoneJung Yoon
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Neurosurgery, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Georgios Voloudakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nathan Doran
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Emily Zhang
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christina Dimovasili
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Lei Chen
- Department of Physiology, Spinal Cord and Brain Injury Research Center, University of Kentucky, Lexington, KY, 40536, USA
| | - Zhiping Shao
- Pamela Sklar Division of Psychiatric Genomics, Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Spyros Darmanis
- Departments of Bioengineering and Applied Physics, Stanford University and Chan Zuckerberg Biohub, Stanford, CA, 94305, USA
| | - Cheuk Tang
- Department of Radiology, Neuroscience and Psychiatry Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Jun Tang
- Department of Radiology, Neuroscience and Psychiatry Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Victoria X Wang
- Department of Radiology, Translational and Molecular Imaging Institute at Mount Sinai, New York, NY, USA
| | - Patrick R Hof
- Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Nikolaos K Robakis
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - Anastasios Georgakopoulos
- Center for Molecular Biology and Genetics of Neurodegeneration, Departments of Psychiatry and Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
49
|
Kakarla V, Kaneko N, Nour M, Khatibi K, Elahi F, Liebeskind DS, Hinman JD. Pathophysiologic mechanisms of cerebral endotheliopathy and stroke due to Sars-CoV-2. J Cereb Blood Flow Metab 2021; 41:1179-1192. [PMID: 33530831 PMCID: PMC8142132 DOI: 10.1177/0271678x20985666] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/19/2020] [Accepted: 12/05/2020] [Indexed: 12/15/2022]
Abstract
Cerebrovascular events have emerged as a central feature of the clinical syndrome associated with Sars-CoV-2 infection. This increase in infection-related strokes is marked by atypical presentations including stroke in younger patients and a high rate of hemorrhagic transformation after ischemia. A variety of pathogenic mechanisms may underlie this connection. Efforts to identify synergism in the pathophysiology underlying stroke and Sars-CoV-2 infection can inform the understanding of both conditions in novel ways. In this review, the molecular cascades connected to Sars-CoV-2 infection are placed in the context of the cerebral vasculature and in relationship to pathways known to be associated with stroke. Cytokine-mediated promotion of systemic hypercoagulability is suggested while direct Sars-CoV-2 infection of cerebral endothelial cells may also contribute. Endotheliopathy resulting from direct Sars-CoV-2 infection of the cerebral vasculature can modulate ACE2/AT1R/MasR signaling pathways, trigger direct viral activation of the complement cascade, and activate feed-forward cytokine cascades that impact the blood-brain barrier. All of these pathways are already implicated as independent mechanisms driving stroke and cerebrovascular injury irrespective of Sars-CoV-2. Recognizing the overlap of molecular pathways triggered by Sars-CoV-2 infection with those implicated in the pathogenesis of stroke provides an opportunity to identify future therapeutics targeting both Sars-CoV-2 and stroke thereby reducing the impact of the global pandemic.
Collapse
Affiliation(s)
- Visesha Kakarla
- School of Medicine, University of California San Diego, San Diego, CA, USA
| | - Naoki Kaneko
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - May Nour
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kasra Khatibi
- Department of Radiological Sciences, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Fanny Elahi
- Memory and Aging Center, Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA, USA
| | - David S Liebeskind
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Jason D Hinman
- Department of Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
50
|
Kummer KK, Zeidler M, Kalpachidou T, Kress M. Role of IL-6 in the regulation of neuronal development, survival and function. Cytokine 2021; 144:155582. [PMID: 34058569 DOI: 10.1016/j.cyto.2021.155582] [Citation(s) in RCA: 103] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/17/2022]
Abstract
The pleiotropic cytokine interleukin-6 (IL-6) is emerging as a molecule with both beneficial and destructive potentials. It can exert opposing actions triggering either neuron survival after injury or causing neurodegeneration and cell death in neurodegenerative or neuropathic disorders. Importantly, neurons respond differently to IL-6 and this critically depends on their environment and whether they are located in the peripheral or the central nervous system. In addition to its hub regulator role in inflammation, IL-6 is recently emerging as an important regulator of neuron function in health and disease, offering exciting possibilities for more mechanistic insight into the pathogenesis of mental, neurodegenerative and pain disorders and for developing novel therapies for diseases with neuroimmune and neurogenic pathogenic components.
Collapse
Affiliation(s)
- Kai K Kummer
- Institute of Physiology, Medical University of Innsbruck, Austria
| | | | | | - Michaela Kress
- Institute of Physiology, Medical University of Innsbruck, Austria.
| |
Collapse
|