1
|
Liu J, Qing T, He M, Xu L, Wu Z, Huang M, Liu Z, Zhang Y, Li Z, Yang W, Liu J, Li J. Transcriptomics, single-cell sequencing and spatial sequencing-based studies of cerebral ischemia. Eur J Med Res 2025; 30:326. [PMID: 40275374 PMCID: PMC12020253 DOI: 10.1186/s40001-025-02596-2] [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: 02/21/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
With high disability and mortality rate as well as highly complex pathogenesis, cerebral ischemia is highly morbid, prone to recurrence. To comprehensively understand the pathophysiological process of cerebral ischemia and to find new therapeutic strategies, a new approach to cerebral ischemia transcriptomics has emerged in recent years. By integrating data from multiple levels of transcriptomics, such as transcriptomics, single-cell transcriptomics, and spatial transcriptomics, this new approach can provide powerful help in revealing the molecular mechanisms of cerebral ischemia occurrence and development. Key findings highlight the critical roles of inflammation, blood-brain barrier dysfunction, and mitochondrial dysregulation in cerebral ischemia, offering potential biomarkers and therapeutic targets for early diagnosis and personalized treatment. A review of the research progress of cerebral ischemic injury mechanism under the analysis of the comprehensive transcriptomics research method was presented in this article, aiming to study the potential mechanism to provide new, innovative therapeutic strategies for this disease.
Collapse
Affiliation(s)
- Jiaming Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Tao Qing
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Mei He
- Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory of Birth Defects Research and Prevention, Changsha, Hunan, China
| | - Liu Xu
- International Education School, Hunan University of Medicine, Huaihua, Hunan, China
| | - Zhuxiang Wu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Meiting Huang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Zheyu Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Ye Zhang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Zisheng Li
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Wenhui Yang
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Junbo Liu
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China
| | - Jie Li
- Basic Medical College of Hunan University of Medicine, Huaihua, Hunan, China.
- Huaihua Key Laboratory of Ion Channels and Complex Diseases, Huaihua, Hunan, China.
| |
Collapse
|
2
|
Wang Y, Yuan T, Lyu T, Zhang L, Wang M, He Z, Wang Y, Li Z. Mechanism of inflammatory response and therapeutic effects of stem cells in ischemic stroke: current evidence and future perspectives. Neural Regen Res 2025; 20:67-81. [PMID: 38767477 PMCID: PMC11246135 DOI: 10.4103/1673-5374.393104] [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: 07/18/2023] [Revised: 10/13/2023] [Accepted: 11/21/2023] [Indexed: 05/22/2024] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide, with an increasing trend and tendency for onset at a younger age. China, in particular, bears a high burden of stroke cases. In recent years, the inflammatory response after stroke has become a research hotspot: understanding the role of inflammatory response in tissue damage and repair following ischemic stroke is an important direction for its treatment. This review summarizes several major cells involved in the inflammatory response following ischemic stroke, including microglia, neutrophils, monocytes, lymphocytes, and astrocytes. Additionally, we have also highlighted the recent progress in various treatments for ischemic stroke, particularly in the field of stem cell therapy. Overall, understanding the complex interactions between inflammation and ischemic stroke can provide valuable insights for developing treatment strategies and improving patient outcomes. Stem cell therapy may potentially become an important component of ischemic stroke treatment.
Collapse
Affiliation(s)
- Yubo Wang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Tingli Yuan
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
| | - Tianjie Lyu
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Meng Wang
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Zhiying He
- Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai, China
- Institute for Regenerative Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yongjun Wang
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| | - Zixiao Li
- Vascular Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- China National Clinical Research Center for Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- National Center for Healthcare Quality Management in Neurological Diseases, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
- Research Unit of Artificial Intelligence in Cerebrovascular Disease, Chinese Academy of Medical Sciences, Beijing, China
- Beijing Engineering Research Center of Digital Healthcare for Neurological Diseases, Beijing, China
| |
Collapse
|
3
|
Ritzel RM, Jiang D, McCullough LD. CAMs in command: aging brain macrophages fine-tune stroke immune responses. Trends Neurosci 2024; 47:965-967. [PMID: 39443199 PMCID: PMC11631633 DOI: 10.1016/j.tins.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/25/2024]
Abstract
Central nervous system-associated macrophages (CAMs) are a unique subset of immune cells located at the interface between the blood and the brain parenchyma. In a recent study in mice, Levard and colleagues found that CAMs regulate immune cell trafficking, endothelial activation, and antigen presentation following stroke exclusively in aged animals, underscoring the importance of using translationally relevant models for studying age-related diseases.
Collapse
Affiliation(s)
- Rodney M Ritzel
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Danye Jiang
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
4
|
Levard D, Seillier C, Bellemain-Sagnard M, Fournier AP, Lemarchand E, Dembech C, Riou G, McDade K, Smith C, McQuaid C, Montagne A, Amann L, Prinz M, Vivien D, Rubio M. Central nervous system-associated macrophages modulate the immune response following stroke in aged mice. Nat Neurosci 2024; 27:1721-1733. [PMID: 38961228 DOI: 10.1038/s41593-024-01695-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
Age is a major nonmodifiable risk factor for ischemic stroke. Central nervous system-associated macrophages (CAMs) are resident immune cells located along the brain vasculature at the interface between the blood circulation and the parenchyma. By using a clinically relevant thromboembolic stroke model in young and aged male mice and corresponding human tissue samples, we show that during aging, CAMs acquire a central role in orchestrating immune cell trafficking after stroke through the specific modulation of adhesion molecules by endothelial cells. The absence of CAMs provokes increased leukocyte infiltration (neutrophils and CD4+ and CD8+ T lymphocytes) and neurological dysfunction after stroke exclusively in aged mice. Major histocompatibility complex class II, overexpressed by CAMs during aging, plays a significant role in the modulation of immune responses to stroke. We demonstrate that during aging, CAMs become central coordinators of the neuroimmune response that ensure a long-term fine-tuning of the immune responses triggered by stroke.
Collapse
Affiliation(s)
- Damien Levard
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Célia Seillier
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Mathys Bellemain-Sagnard
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Antoine Philippe Fournier
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Eloïse Lemarchand
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Chantal Dembech
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France
| | - Gaëtan Riou
- INSERM U1234 'PAn'THER', Flow Cytometry Core-IRIB, Rouen, France
| | - Karina McDade
- Academic Neuropathology, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Colin Smith
- Academic Neuropathology, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Conor McQuaid
- Dementia Research Institute at the University of Edinburgh, Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh Medical School, Edinburgh, UK
| | - Axel Montagne
- Dementia Research Institute at the University of Edinburgh, Centre for Clinical Brain Sciences, Chancellor's Building, Edinburgh Medical School, Edinburgh, UK
| | - Lukas Amann
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
| | - Marco Prinz
- Institute of Neuropathology, Medical Faculty, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Denis Vivien
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France.
- Department of Clinical Research, Caen-Normandie University Hospital, CHU, Caen, France.
| | - Marina Rubio
- Normandie University, UNICAEN, Université Caen Normandie, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders (PhIND), GIP Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), Caen, France.
| |
Collapse
|
5
|
Wang K, Yang L, Li Q, Yang X, Chen Z, Zhou Y, Jia Y, Gong Z. Long-Term Alcohol Exposure Aggravates Ischemic Stroke-Induced Damage by Promoting Pericyte NLRP3 Inflammasome Activation via Pre-Activating the TLR4/NF-κB Pathway in Rats. J Inflamm Res 2024; 17:4791-4810. [PMID: 39051052 PMCID: PMC11268848 DOI: 10.2147/jir.s465780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/25/2024] [Indexed: 07/27/2024] Open
Abstract
Background Ischemic stroke (IS) is one of the leading causes of death and disability in the world, and alcohol consumption has been gaining attention as an independent risk factor for IS. Blood-brain barrier (BBB) dysfunction and neuroinflammation are the core of cerebral ischemia/reperfusion (I/R) injury, and pericytes play a crucial role in the structure and function. This study is to explore the effects of long-term alcohol consumption on IS and the potential mechanisms of pericytes. Methods Rat models of long-term alcohol intake followed by transient middle cerebral artery occlusion stroke (EtOH+tMCAO) and cell models of oxygen-glucose deprivation/reoxygenation (OGD/R) with alcohol pre-treatment were constructed. Results Worsened infarct volume, neurological scores, and BBB disruption were observed in the EtOH+tMCAO group compared with the tMCAO group, and immunofluorescence staining showed increased pericytes NLPR3 inflammasome activation at the ischemic penumbra. In vitro, pericyte mortality and LDH release elevated pre-treated by alcohol after OGD/R, and amplified expression of NLRP3 inflammasome was detected by Western blotting and qPCR. Alcohol pre-treatment activated the TLR4/NF-κB pathway, and transfecting pericytes with TLR4-small interfering RNA (siRNA) to block TLR4 signaling markedly restrained NLRP3 inflammasome over-activation. Injecting TAK-242 in rats alleviated neurological impairment caused by alcohol. Conclusion Long-term alcohol pre-treatment aggravated ischemic stroke-induced brain damage by activating NLRP3 inflammasome via TLR4/NF-κB signaling pathway in the pericytes.
Collapse
Affiliation(s)
- Kaixin Wang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Lingfei Yang
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Qingsheng Li
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Xuan Yang
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Ziyi Chen
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Yongyan Zhou
- Department of Electrocardiogram, Zhoukou Center Hospital, Zhoukou, People’s Republic of China
| | - Yanjie Jia
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| | - Zhe Gong
- Department of Neurology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, People’s Republic of China
| |
Collapse
|
6
|
Guan J, Gan L, Yan C, Hou B, Fan Y. Vitamin D deficiency and increased inflammatory factor intercellular cell adhesion molecule-1 indicate severe leukoaraiosis in northern China. FRIGID ZONE MEDICINE 2024; 4:102-109. [DOI: 10.1515/fzm-2024-0011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2024]
Abstract
Abstract
Background and objective
Commonly plaguing in the frigid zone of the world, vitamin D deficiency, as indicated by low levels of 25-hydroxyvitamin D, exacerbated inflammatory responses and impaired endothelial function. Leukoaraiosis (LA) is a prevalent cause of cognitive dysfunction in the elderly and is potentially associated with inflammatory responses. This study aimed to investigate the impact of vitamin D on the severity of LA.
Methods
Patients with LA were categorized based on 3.0 T brain MRI findings into mild (N = 43), moderate (N = 40), or severe groups (N = 29) using the Fazekas scale (scoring 1-6). A control group consisting of 41 healthy individuals was included. Serum fibrinogen C, homocysteine, plasma 25-hydroxyvitamin D, and intercellular cell adhesion molecule-1 (ICAM-1) levels were measured using ELISA.
Results
All LA severity groups exhibited lower plasma 25-hydroxyvitamin D levels compared to the control group, with a more pronounced decrease observed as LA severity increased. Low plasma 25-hydroxyvitamin D was identified as an independent risk factor for LA (P < 0.05) according to Multiple logistic regression analysis. Additionally, a negative association was observed between 25-hydroxyvitamin D and vascular inflammatory factor ICAM-1.
Conclusions
Disease severity positively correlated with levels of the inflammatory marker ICAM-1, worsening as plasma 25-hydroxyvitamin D concentration decreased. Low 25-hydroxyvitamin D emerged as an independent risk factor for LA, potentially exacerbating the inflammatory response. These findings suggest 25-hydroxyvitamin D supplementation as a potential therapeutic approach for LA.
Collapse
Affiliation(s)
- Jiaxin Guan
- Second Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Lu Gan
- Second Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Chaoqi Yan
- The Physical examination center of the Second Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Boyu Hou
- Second Affiliated Hospital of Harbin Medical University , Harbin , China
| | - Ying Fan
- Second Affiliated Hospital of Harbin Medical University , Harbin , China
| |
Collapse
|
7
|
Planas AM. Role of microglia in stroke. Glia 2024; 72:1016-1053. [PMID: 38173414 DOI: 10.1002/glia.24501] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 12/07/2023] [Accepted: 12/19/2023] [Indexed: 01/05/2024]
Abstract
Microglia play key roles in the post-ischemic inflammatory response and damaged tissue removal reacting rapidly to the disturbances caused by ischemia and working to restore the lost homeostasis. However, the modified environment, encompassing ionic imbalances, disruption of crucial neuron-microglia interactions, spreading depolarization, and generation of danger signals from necrotic neurons, induce morphological and phenotypic shifts in microglia. This leads them to adopt a proinflammatory profile and heighten their phagocytic activity. From day three post-ischemia, macrophages infiltrate the necrotic core while microglia amass at the periphery. Further, inflammation prompts a metabolic shift favoring glycolysis, the pentose-phosphate shunt, and lipid synthesis. These shifts, combined with phagocytic lipid intake, drive lipid droplet biogenesis, fuel anabolism, and enable microglia proliferation. Proliferating microglia release trophic factors contributing to protection and repair. However, some microglia accumulate lipids persistently and transform into dysfunctional and potentially harmful foam cells. Studies also showed microglia that either display impaired apoptotic cell clearance, or eliminate synapses, viable neurons, or endothelial cells. Yet, it will be essential to elucidate the viability of engulfed cells, the features of the local environment, the extent of tissue damage, and the temporal sequence. Ischemia provides a rich variety of region- and injury-dependent stimuli for microglia, evolving with time and generating distinct microglia phenotypes including those exhibiting proinflammatory or dysfunctional traits and others showing pro-repair features. Accurate profiling of microglia phenotypes, alongside with a more precise understanding of the associated post-ischemic tissue conditions, is a necessary step to serve as the potential foundation for focused interventions in human stroke.
Collapse
Affiliation(s)
- Anna M Planas
- Cerebrovascular Research Laboratory, Department of Neuroscience and Experimental Therapeutics, Instituto de Investigaciones Biomédicas de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Cerebrovascular Diseases, Area of Clinical and Experimental Neuroscience, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS)-Hospital Clínic, Barcelona, Spain
| |
Collapse
|
8
|
Kaur J, Boyd ED, Ding G, Zhang L, Luo H, Li Q, Li L, Wei M, Landschoot-Ward J, Chopp M, Zhang Z, Jiang Q. The Association between Glymphatic System and Perivascular Macrophages in Brain Waste Clearance. Diagnostics (Basel) 2024; 14:731. [PMID: 38611644 PMCID: PMC11011895 DOI: 10.3390/diagnostics14070731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/13/2024] [Accepted: 03/14/2024] [Indexed: 04/14/2024] Open
Abstract
The glymphatic system suggests the convective bulk flow of cerebrospinal fluid (CSF) through perivascular spaces and the interstitial spaces of the brain parenchyma for the rapid removal of toxic waste solutes from the brain. However, the presence of convective bulk flow within the brain interstitial spaces is still under debate. We first addressed this argument to determine the involvement of the glymphatic system in brain waste clearance utilizing contrast-enhanced 3D T1-weighted imaging (T1WI), diffusion tensor imaging (DTI), and confocal microscopy imaging. Furthermore, perivascular macrophages (PVMs), which are immune cells located within perivascular spaces, have not been thoroughly explored for their association with the glymphatic system. Therefore, we investigated tracer uptake by PVMs in the perivascular spaces of both the arteries/arterioles and veins/venules and the potential association of PVMs in assisting the glymphatic system for interstitial waste clearance. Our findings demonstrated that both convective bulk flow and diffusion are responsible for the clearance of interstitial waste solutes from the brain parenchyma. Furthermore, our results suggested that PVMs may play an important function in glymphatic system-mediated interstitial waste clearance. The glymphatic system and PVMs could be targeted to enhance interstitial waste clearance in patients with waste-associated neurological conditions and aging.
Collapse
Affiliation(s)
- Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
| | - Edward D. Boyd
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
| | - Hao Luo
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Min Wei
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Julie Landschoot-Ward
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
| | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI 48202, USA; (J.K.); (E.D.B.); (G.D.); (L.Z.); (H.L.); (Q.L.); (L.L.); (M.W.); (J.L.-W.); (M.C.); (Z.Z.)
- Department of Physics, Oakland University, Rochester, MI 48309, USA
- Department of Radiology, Michigan State University, East Lansing, MI 48824, USA
- Department of Neurology, Wayne State University, Detroit, MI 48202, USA
| |
Collapse
|
9
|
Glavan M, Jelic A, Levard D, Frösen J, Keränen S, Franx BAA, Bras AR, Louet ER, Dénes Á, Merlini M, Vivien D, Rubio M. CNS-associated macrophages contribute to intracerebral aneurysm pathophysiology. Acta Neuropathol Commun 2024; 12:43. [PMID: 38500201 PMCID: PMC10946177 DOI: 10.1186/s40478-024-01756-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2023] [Accepted: 03/06/2024] [Indexed: 03/20/2024] Open
Abstract
Intracerebral aneurysms (IAs) are pathological dilatations of cerebral arteries whose rupture leads to subarachnoid hemorrhage, a significant cause of disability and death. Inflammation is recognized as a critical contributor to the formation, growth, and rupture of IAs; however, its precise actors have not yet been fully elucidated. Here, we report CNS-associated macrophages (CAMs), also known as border-associated macrophages, as one of the key players in IA pathogenesis, acting as critical mediators of inflammatory processes related to IA ruptures. Using a new mouse model of middle cerebral artery (MCA) aneurysms we show that CAMs accumulate in the IA walls. This finding was confirmed in a human MCA aneurysm obtained after surgical clipping, together with other pathological characteristics found in the experimental model including morphological changes and inflammatory cell infiltration. In addition, in vivo longitudinal molecular MRI studies revealed vascular inflammation strongly associated with the aneurysm area, i.e., high expression of VCAM-1 and P-selectin adhesion molecules, which precedes and predicts the bleeding extent in the case of IA rupture. Specific CAM depletion by intracerebroventricular injection of clodronate liposomes prior to IA induction reduced IA formation and rupture rate. Moreover, the absence of CAMs ameliorated the outcome severity of IA ruptures resulting in smaller hemorrhages, accompanied by reduced neutrophil infiltration. Our data shed light on the unexplored role of CAMs as main actors orchestrating the progression of IAs towards a rupture-prone state.
Collapse
Affiliation(s)
- Martina Glavan
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France
- Department of Neuroscience, Yale School of Medicine, Yale University, 333 Cedar Street, New Haven, CT, 06510, USA
| | - Ana Jelic
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France
| | - Damien Levard
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France
| | - Juhana Frösen
- Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital and AIV Institute for Molecular Medicine, University of Eastern Finland, Kuopio, Finland
- Dept of Neurosurgery, Tampere University Hospital and Hemorrhagic Brain Pathology Research Group, Tampere University, Tampere, Finland
| | - Sara Keränen
- Hemorrhagic Brain Pathology Research Group, Kuopio University Hospital and AIV Institute for Molecular Medicine, University of Eastern Finland, Kuopio, Finland
- Dept of Neurosurgery, Tampere University Hospital and Hemorrhagic Brain Pathology Research Group, Tampere University, Tampere, Finland
| | - Bart A A Franx
- Translational Neuroimaging Group, Center for Image Sciences, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Ana-Rita Bras
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
- János Szentágothai Doctoral School of Neurosciences, Schools of PhD Studies, Semmelweis University, Budapest, Hungary
| | - Estelle R Louet
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France
| | - Ádám Dénes
- "Momentum" Laboratory of Neuroimmunology, Institute of Experimental Medicine, Budapest, Hungary
| | - Mario Merlini
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France
| | - Denis Vivien
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France
- Department of Clinical Research, Caen Normandie University Hospital, Caen, France
| | - Marina Rubio
- UNICAEN, INSERM U1237, Etablissement Français du Sang, Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), PHIND Boulevard Henri Becquerel, Normandie University, 14000, Caen Cedex, Caen, France.
| |
Collapse
|
10
|
Lebrun F, Levard D, Lemarchand E, Yetim M, Furon J, Potzeha F, Marie P, Lesept F, Blanc M, Haelewyn B, Rubio M, Letourneur A, Violle N, Orset C, Vivien D. Improving stroke outcomes in hyperglycemic mice by modulating tPA/NMDAR signaling to reduce inflammation and hemorrhages. Blood Adv 2024; 8:1330-1344. [PMID: 38190586 PMCID: PMC10943589 DOI: 10.1182/bloodadvances.2023011744] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 12/15/2023] [Accepted: 12/17/2023] [Indexed: 01/10/2024] Open
Abstract
ABSTRACT The pharmacological intervention for ischemic stroke hinges on intravenous administration of the recombinant tissue-type plasminogen activator (rtPA, Alteplase/Actilyse) either as a standalone treatment or in conjunction with thrombectomy. However, despite its clinical significance, broader use of rtPA is constrained because of the risk of hemorrhagic transformations (HTs). Furthermore, the presence of diabetes or chronic hyperglycemia is associated with an elevated risk of HT subsequent to thrombolysis. This detrimental impact of tPA on the neurovascular unit in patients with hyperglycemia has been ascribed to its capacity to induce endothelial N-methyl-D-aspartate receptor (NMDAR) signaling, contributing to compromised blood-brain barrier integrity and neuroinflammatory processes. In a mouse model of thromboembolic stroke with chronic hyperglycemia, we assessed the effectiveness of rtPA and N-acetylcysteine (NAC) as thrombolytic agents. We also tested the effect of blocking tPA/NMDAR signaling using a monoclonal antibody, Glunomab. Magnetic resonance imaging, speckle contrast imaging, flow cytometry, and behavioral tasks were used to evaluate stroke outcomes. In hyperglycemic animals, treatment with rtPA resulted in lower recanalization rates and increased HTs. Conversely, NAC treatment reduced lesion sizes while mitigating HTs. After a single administration, either in standalone or combined with rtPA-induced thrombolysis, Glunomab reduced brain lesion volumes, HTs, and neuroinflammation after stroke, translating into improved neurological outcomes. Additionally, we demonstrated the therapeutic efficacy of Glunomab in combination with NAC or as a standalone strategy in chronic hyperglycemic animals. Counteracting tPA-dependent endothelial NMDAR signaling limits ischemic damages induced by both endogenous and exogenous tPA, including HTs and inflammatory processes after ischemic stroke in hyperglycemic animals.
Collapse
Affiliation(s)
- Florent Lebrun
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
- STROK@LLIANCE, ETAP-Lab, Caen, France
| | - Damien Levard
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | - Eloïse Lemarchand
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | - Mervé Yetim
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | - Jonathane Furon
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | - Fanny Potzeha
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | - Pauline Marie
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | | | | | - Benoit Haelewyn
- GIP Cyceron, Caen, France
- Experimental Stroke Research Platform, Normandie University, CURB, Caen, France
| | - Marina Rubio
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
| | | | | | - Cyrille Orset
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
- Experimental Stroke Research Platform, Normandie University, CURB, Caen, France
| | - Denis Vivien
- Normandie University, UNICAEN, INSERM UMR-S U1237, Physiopathology and Imaging of Neurological Disorders, GIP Cyceron, Institute Blood and Brain @ Caen-Normandie, Caen, France
- Experimental Stroke Research Platform, Normandie University, CURB, Caen, France
- Department of Clinical Research, Caen-Normandie University Hospital, Caen, France
| |
Collapse
|
11
|
Wen W, Cheng J, Tang Y. Brain perivascular macrophages: current understanding and future prospects. Brain 2024; 147:39-55. [PMID: 37691438 PMCID: PMC10766266 DOI: 10.1093/brain/awad304] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 07/28/2023] [Accepted: 08/07/2023] [Indexed: 09/12/2023] Open
Abstract
Brain perivascular macrophages are specialized populations of macrophages that reside in the space around cerebral vessels, such as penetrating arteries and venules. With the help of cutting-edge technologies, such as cell fate mapping and single-cell multi-omics, their multifaceted, pivotal roles in phagocytosis, antigen presentation, vascular integrity maintenance and metabolic regulation have more recently been further revealed under physiological conditions. Accumulating evidence also implies that perivascular macrophages are involved in the pathogenesis of neurodegenerative disease, cerebrovascular dysfunction, autoimmune disease, traumatic brain injury and epilepsy. They can act in either protective or detrimental ways depending on the disease course and stage. However, the underlying mechanisms of perivascular macrophages remain largely unknown. Therefore, we highlight potential future directions in research on perivascular macrophages, including the utilization of genetic mice and novel therapeutic strategies that target these unique immune cells for neuroprotective purposes. In conclusion, this review provides a comprehensive update on the current knowledge of brain perivascular macrophages, shedding light on their pivotal roles in central nervous system health and disease.
Collapse
Affiliation(s)
- Wenjie Wen
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Jinping Cheng
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| | - Yamei Tang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Brain Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
- Nanhai Translational Innovation Center of Precision Immunology, Sun Yat-sen Memorial Hospital, Foshan 528200, China
| |
Collapse
|
12
|
Kaur J, Boyd E, Ding G, Zhang L, Luo H, Li Q, Li L, Wei M, Landschoot-Ward J, Chopp M, Zhang Z, Jiang Q. The Association between Glymphatic System and Perivascular Macrophages in Brain Waste Clearance. RESEARCH SQUARE 2023:rs.3.rs-3390074. [PMID: 37886481 PMCID: PMC10602168 DOI: 10.21203/rs.3.rs-3390074/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The glymphatic system suggests the convective bulk flow of cerebrospinal fluid (CSF) through perivascular spaces and the interstitial spaces of the brain parenchyma for the rapid removal of toxic waste solutes from the brain. However, the presence of convective bulk flow within the brain interstitial spaces is still under debate. We first addressed this argument to determine the involvement of the glymphatic system in brain waste clearance utilizing contrast-enhanced 3D T1-weighted imaging (T1WI), diffusion tensor imaging (DTI), and confocal microscopy imaging. Furthermore, perivascular macrophages (PVMs), which are immune cells located within perivascular spaces, have not been thoroughly explored for their association with the glymphatic system. Therefore, we investigated tracer uptake by PVMs in the perivascular spaces of both the arteries/arterioles and veins/venules and the potential association of PVMs in assisting the glymphatic system for interstitial waste clearance. Our findings demonstrated that both convective bulk flow and diffusion are responsible for the clearance of interstitial waste solutes from the brain parenchyma. Furthermore, our results suggested that PVMs play an important function in glymphatic system-mediated interstitial waste clearance. The glymphatic system and PVMs could be targeted to enhance interstitial waste clearance in patients with waste-associated neurological conditions and aging.
Collapse
Affiliation(s)
- Jasleen Kaur
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
| | - Edward Boyd
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Radiology, Michigan State University, Lasing, MI, USA
| | - Guangliang Ding
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Radiology, Michigan State University, Lasing, MI, USA
| | - Li Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Physiology, Michigan State University, Lasing, MI, USA
| | - Hao Luo
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Qingjiang Li
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Lian Li
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | - Min Wei
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
| | | | - Michael Chopp
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
- Department of Physiology, Michigan State University, Lasing, MI, USA
- Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Zhenggang Zhang
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Physiology, Michigan State University, Lasing, MI, USA
- Department of Neurology, Wayne State University, Detroit, MI, USA
| | - Quan Jiang
- Department of Neurology, Henry Ford Health System, Detroit, MI, USA
- Department of Physics, Oakland University, Rochester, MI, USA
- Department of Radiology, Michigan State University, Lasing, MI, USA
- Department of Neurology, Wayne State University, Detroit, MI, USA
| |
Collapse
|
13
|
Goertz JE, Garcia-Bonilla L, Iadecola C, Anrather J. Immune compartments at the brain's borders in health and neurovascular diseases. Semin Immunopathol 2023; 45:437-449. [PMID: 37138042 PMCID: PMC10279585 DOI: 10.1007/s00281-023-00992-6] [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: 02/07/2023] [Accepted: 03/14/2023] [Indexed: 05/05/2023]
Abstract
Recent evidence implicates cranial border immune compartments in the meninges, choroid plexus, circumventricular organs, and skull bone marrow in several neuroinflammatory and neoplastic diseases. Their pathogenic importance has also been described for cardiovascular diseases such as hypertension and stroke. In this review, we will examine the cellular composition of these cranial border immune niches, the potential pathways through which they might interact, and the evidence linking them to cardiovascular disease.
Collapse
Affiliation(s)
- Jennifer E Goertz
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA
| | - Lidia Garcia-Bonilla
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA
| | - Josef Anrather
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61St Street; RR-405, New York, NY, 10065, USA.
| |
Collapse
|
14
|
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
|
15
|
Árokszállási T, Balogh E, Orbán-Kálmándi R, Pásztor M, Árokszállási A, Nagy EB, Belán I, May Z, Csépány T, Csiba L, Bagoly Z, Oláh L. Elevated Blood Alcohol Concentration Is Associated with Improved Clinical Outcomes of Intravenous Thrombolysis Treatment in Acute Ischemic Stroke Patients—A Retrospective Study. J Clin Med 2023; 12:jcm12062238. [PMID: 36983239 PMCID: PMC10051873 DOI: 10.3390/jcm12062238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 03/04/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023] Open
Abstract
Background: Intravenous thrombolysis (IVT) improves acute ischemic stroke (AIS) outcomes, but with limited success. In addition, ethanol potentiates the effect of r-tPA in ischemia models. Methods: The effect of acute alcohol consumption on IVT outcomes was investigated in a retrospective cohort study. AIS patients with detectable blood alcohol concentration (BAC) during IVT were included (alcohol group; n = 60). For each case, 3 control subjects who underwent IVT but denied alcohol consumption were matched in terms of age, sex, affected brain area, and stroke severity. Outcomes were determined using the NIHSS at 7 days and the modified Rankin scale (mRS) at 90 days. Results: Patients were younger and had a less severe stroke than in a standard stroke study. Favorable long-term outcomes (mRS 0–2) occurred significantly more frequently in the alcohol group compared to controls (90% vs. 63%, p < 0.001). However, the rates of hemorrhagic transformation were similar. Multiple logistic regression models identified elevated BAC as a significant protective factor against unfavorable short-term (OR: 0.091, 95% CI: 0.036–0.227, p < 0.001) and long-term outcomes (OR: 0.187, 95% CI: 0.066–0.535, p = 0.002). In patients with BAC > 0.2%, significantly lower NIHSS was observed at 3 and 7 days after IVT vs. in those with 0.01–0.2% ethanol levels. Conclusion: Elevated BAC is associated with improved outcomes in IVT-treated AIS without affecting safety.
Collapse
Affiliation(s)
- Tamás Árokszállási
- Department of Neurology, Faculty of Medicine, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
| | - Eszter Balogh
- Department of Neurology, Faculty of Medicine, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
| | - Rita Orbán-Kálmándi
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, H-4032 Debrecen, Hungary
| | - Máté Pásztor
- Department of Neurology, Medical Centre, Hungarian Defence Forces, 44 Károly Róbert krt, H-1134 Budapest, Hungary
| | - Anita Árokszállási
- Department of Oncology, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, H-4032 Debrecen, Hungary
| | - Edit Boglárka Nagy
- Division of Radiology and Imaging Science, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, H-4032 Debrecen, Hungary
| | - Ivett Belán
- Division of Radiology and Imaging Science, Department of Medical Imaging, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, H-4032 Debrecen, Hungary
| | - Zsolt May
- Department of Neurology, Medical Centre, Hungarian Defence Forces, 44 Károly Róbert krt, H-1134 Budapest, Hungary
| | - Tünde Csépány
- Department of Neurology, Faculty of Medicine, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
| | - László Csiba
- Department of Neurology, Faculty of Medicine, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
- ELKH-DE Cerebrovascular Research Group, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
| | - Zsuzsa Bagoly
- Division of Clinical Laboratory Sciences, Department of Laboratory Medicine, Faculty of Medicine, University of Debrecen, 98 Nagyerdei krt, H-4032 Debrecen, Hungary
- ELKH-DE Cerebrovascular Research Group, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
| | - László Oláh
- Department of Neurology, Faculty of Medicine, Doctoral School of Neuroscience, University of Debrecen, 22 Móricz Zsigmond krt, H-4032 Debrecen, Hungary
- Correspondence: ; Tel.: +36-52-255-341; Fax: +36-52-255-590
| |
Collapse
|
16
|
Innate immune tolerance against adolescent intermittent alcohol exposure-induced behavioral abnormalities in adult mice. Int Immunopharmacol 2022; 113:109250. [DOI: 10.1016/j.intimp.2022.109250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 08/23/2022] [Accepted: 09/09/2022] [Indexed: 11/05/2022]
|
17
|
Gerganova G, Riddell A, Miller AA. CNS border-associated macrophages in the homeostatic and ischaemic brain. Pharmacol Ther 2022; 240:108220. [PMID: 35667516 DOI: 10.1016/j.pharmthera.2022.108220] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Revised: 05/17/2022] [Accepted: 05/31/2022] [Indexed: 12/14/2022]
Abstract
CNS border-associated macrophages (BAMs) are a small population of specialised macrophages localised in the choroid plexus, meningeal and perivascular spaces. Until recently, the function of this elusive cell type was poorly understood and largely overlooked, especially in comparison to microglia, the primary brain resident immune cell. However, the recent single cell immunophenotyping or transcriptomic analysis of BAM subsets in the homeostatic brain, coupled with the rapid emergence of new studies exploring BAM functions in various cerebral pathologies, including Alzheimer's disease, hypertension-induced neurovascular and cognitive dysfunction, and ischaemic stroke, has unveiled previously unrecognised heterogeneity and spatial-temporal complexity in BAM populations as well as their contributions to brain homeostasis and disease. In this review, we discuss the implications of this new-found knowledge on our current understanding of BAM function in ischaemic stroke. We first provide a comprehensive overview and discussion of the cell-surface expression profiles, transcriptional signatures and potential functional phenotypes of homeostatic BAM subsets described in recent studies. Evidence for their putative physiological roles is examined, including their involvement in immunological surveillance, waste clearance, and vascular permeability. We discuss the evidence supporting the accumulation and genetic transformation of BAMs in response to ischaemia and appraise the experimental evidence that BAM function might be deleterious in the acute phase of stroke, while considering the mechanisms by which BAMs may influence stroke outcomes in the longer term. Finally, we review the therapeutic potential of immunomodulatory strategies as an approach to stroke management, highlighting current challenges in the field and key issues relating to BAMs, and how BAMs could be harnessed experimentally to support future translational research.
Collapse
Affiliation(s)
- Gabriela Gerganova
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Alexandra Riddell
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom
| | - Alyson A Miller
- British Heart Foundation Glasgow Cardiovascular Research Centre, Institute of Cardiovascular & Medical Sciences, University of Glasgow, Glasgow G12 8TA, United Kingdom.
| |
Collapse
|
18
|
Zheng L, Guo Y, Zhai X, Zhang Y, Chen W, Zhu Z, Xuan W, Li P. Perivascular macrophages in the CNS: From health to neurovascular diseases. CNS Neurosci Ther 2022; 28:1908-1920. [PMID: 36128654 PMCID: PMC9627394 DOI: 10.1111/cns.13954] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 07/15/2022] [Accepted: 07/22/2022] [Indexed: 02/06/2023] Open
Abstract
Brain perivascular macrophages (PVMs) are attracting increasing attention as this emerging cell population in the brain has multifaced roles in supporting the central nervous system structure, brain development, and maintaining physiological functions. They also widely participate in neurological diseases such as neurodegeneration and ischemic stroke. Moreover, PVMs have been reported to have both beneficial and detrimental effects under different pathological contexts. Advanced research technologies allowed the further in-depth study of PVMs and revealed novel concepts in their origins, differentiation, and regulatory mechanisms. Deepened understanding of the roles of PVMs in different brain pathological conditions can reveal novel phenotypic changes and regulatory signaling, which might pave the way for the development of novel treatment strategies targeting PVMs.
Collapse
Affiliation(s)
- Li Zheng
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yunlu Guo
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaozhu Zhai
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Yueman Zhang
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Weijie Chen
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Ziyu Zhu
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Xuan
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peiying Li
- Department of Anesthesiology, State Key Laboratory of Oncogenes and Related GenesShanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
19
|
Ramos A, Joshi RS, Szabo G. Innate immune activation: Parallels in alcohol use disorder and Alzheimer’s disease. Front Mol Neurosci 2022; 15:910298. [PMID: 36157070 PMCID: PMC9505690 DOI: 10.3389/fnmol.2022.910298] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 07/18/2022] [Indexed: 11/16/2022] Open
Abstract
Alcohol use disorder is associated with systemic inflammation and organ dysfunction especially in the liver and the brain. For more than a decade, studies have highlighted alcohol abuse-mediated impairment of brain function and acceleration of neurodegeneration through inflammatory mechanisms that directly involve innate immune cells. Furthermore, recent studies indicate overlapping genetic risk factors between alcohol use and neurodegenerative disorders, specifically regarding the role of innate immunity in the pathomechanisms of both areas. Considering the pressing need for a better understanding of the relevance of alcohol abuse in dementia progression, here we summarize the molecular mechanisms of neuroinflammation observed in alcohol abuse and Alzheimer’s disease, the most common cause of dementia. In addition, we highlight mechanisms that are already established in the field of Alzheimer’s disease that may be relevant to explore in alcoholism to better understand alcohol mediated neurodegeneration and dementia, including the relevance of the liver-brain axis.
Collapse
Affiliation(s)
- Adriana Ramos
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Radhika S. Joshi
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Gyongyi Szabo
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Broad Institute of MIT and Harvard, Cambridge, MA, United States
- *Correspondence: Gyongyi Szabo,
| |
Collapse
|
20
|
Bigdelou B, Sepand MR, Najafikhoshnoo S, Negrete JAT, Sharaf M, Ho JQ, Sullivan I, Chauhan P, Etter M, Shekarian T, Liang O, Hutter G, Esfandiarpour R, Zanganeh S. COVID-19 and Preexisting Comorbidities: Risks, Synergies, and Clinical Outcomes. Front Immunol 2022; 13:890517. [PMID: 35711466 PMCID: PMC9196863 DOI: 10.3389/fimmu.2022.890517] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/11/2022] [Indexed: 12/15/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its associated symptoms, named coronavirus disease 2019 (COVID-19), have rapidly spread worldwide, resulting in the declaration of a pandemic. When several countries began enacting quarantine and lockdown policies, the pandemic as it is now known truly began. While most patients have minimal symptoms, approximately 20% of verified subjects are suffering from serious medical consequences. Co-existing diseases, such as cardiovascular disease, cancer, diabetes, and others, have been shown to make patients more vulnerable to severe outcomes from COVID-19 by modulating host-viral interactions and immune responses, causing severe infection and mortality. In this review, we outline the putative signaling pathways at the interface of COVID-19 and several diseases, emphasizing the clinical and molecular implications of concurring diseases in COVID-19 clinical outcomes. As evidence is limited on co-existing diseases and COVID-19, most findings are preliminary, and further research is required for optimal management of patients with comorbidities.
Collapse
Affiliation(s)
- Banafsheh Bigdelou
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Mohammad Reza Sepand
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Sahar Najafikhoshnoo
- Department of Electrical Engineering, University of California, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, Irvine, CA, United States
| | - Jorge Alfonso Tavares Negrete
- Department of Electrical Engineering, University of California, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, Irvine, CA, United States
| | - Mohammed Sharaf
- Department of Chemical and Biomolecular Engineering, New York University, New York, NY, United States
| | - Jim Q Ho
- Department of Medicine, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ian Sullivan
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| | - Prashant Chauhan
- Institute of Parasitology, Biology Centre Czech Academy of Science, Ceske Budejovice, Czech Republic
| | - Manina Etter
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Tala Shekarian
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Olin Liang
- Division of Hematology/Oncology, Department of Medicine, Rhode Island Hospital and Warren Alpert Medical School of Brown University, Providence, RI, United States
| | - Gregor Hutter
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Rahim Esfandiarpour
- Department of Electrical Engineering, University of California, Irvine, CA, United States
- Department of Biomedical Engineering, University of California, Irvine, Irvine, CA, United States
- Laboratory for Integrated Nano Bio Electronics Innovation, The Henry Samueli School of Engineering, University of California, Irvine, Irvine, CA, United States
| | - Steven Zanganeh
- Department of Bioengineering, University of Massachusetts Dartmouth, Dartmouth, MA, United States
| |
Collapse
|
21
|
Vore AS, Barney TM, Deak MM, Varlinskaya EI, Deak T. Adolescent intermittent ethanol exposure produces Sex-Specific changes in BBB Permeability: A potential role for VEGFA. Brain Behav Immun 2022; 102:209-223. [PMID: 35245677 PMCID: PMC9277567 DOI: 10.1016/j.bbi.2022.02.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 01/31/2022] [Accepted: 02/27/2022] [Indexed: 12/20/2022] Open
Abstract
Binge drinking that typically begins during adolescence can have long-lasting neurobehavioral consequences, including alterations in the central and peripheral immune systems. Central and peripheral inflammation disrupts blood-brain barrier (BBB) integrity and exacerbates pathology in diseases commonly associated with disturbed BBB function. Thus, the goal of the present studies was to determine long-lasting effects of adolescent intermittent ethanol (AIE) on BBB integrity. For AIE, male and female Sprague Dawley rats were repeatedly exposed to ethanol (4 g/kg, intragastrically) or water during adolescence between postnatal day (P) 30 and P50. In adulthood (∼P75), rats were challenged with fluorescein isothiocyanate (FITC)-tagged Dextran of varying molecular weights (4, 20, & 70 kDa) for assessment of BBB permeability using gross tissue fluorometry (Experiment 1). Experiment 2 extended these effects using immunofluorescence, adding an adult ethanol-exposed group to test for a specific developmental vulnerability. Finally, as a first test of hypothesized mechanism, Experiment 3 examined the effect of AIE on Vascular Endothelial Growth Factor A (VEGFA) and its co-localization with pericytes (identified through expression of platelet derived growth factor receptor beta (PDGFRβ), a key regulatory cell embedded within the BBB. Male, but not female, rats with a history of AIE showed significantly increased dextran permeability in the nucleus accumbens (NAc), cingulate prefrontal cortex (cPFC), and amygdala (AMG). Similar increases in dextran were observed in the hippocampus (HPC) and ventral tegmental area (VTA) of male rats with a history of AIE or equivalent ethanol exposure during adulthood. No changes in BBB permeability were evident in females. When VEGFa expression was examined, male rats exposed to AIE were challenged with 3.5 g/kg ethanol (i.p.) or vehicle acutely in adulthood to assess long-lasting versus acute actions of ethanol. Adult rats with a history of AIE showed significantly fewer total cells expressing VEGFa in the AMG and dHPC following the acute ethanol challenge in adulthood. They also showed a significant reduction in the number of PDGFRβ positive cells that also expressed VEGFa signal. The anatomical distribution of these effects corresponded with increased BBB permeability after AIE (i.e., differential effects in the PVN, AMG, and dHPC). These studies demonstrated sex-specific effects of AIE, with males, but not females, demonstrating long-term increases in BBB permeability that correlated with changes in VEGFa and PDGFRβ protein, two factors known to influence BBB permeability.
Collapse
Affiliation(s)
| | | | | | | | - Terrence Deak
- Developmental Exposure Alcohol Research Center, Behavioral Neuroscience Program, Department of Psychology, Binghamton, NY 13902-6000.
| |
Collapse
|
22
|
Horn AR, Diamond KB, Ng MK, Vakharia RM, Mont MA, Erez O. The Association of Alcohol Use Disorder with Perioperative Complications following Primary Total Hip Arthroplasty. Hip Pelvis 2021; 33:231-238. [PMID: 34938693 PMCID: PMC8654594 DOI: 10.5371/hp.2021.33.4.231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/14/2022] Open
Abstract
Purpose Alcohol use disorder is a leading mental health disorder in the United States. Few studies evaluating the association of alcohol use disorder following primary total hip arthroplasty (THA) have been reported. Therefore, the purpose of this study was to determine whether patients with alcohol use disorder undergoing primary THA have higher rates of: 1) in-hospital lengths of stay (LOS); 2) complications (medical/implant-related); and 3) costs. Materials and Methods Using a nationwide claims database from January 1st, 2005 to March 31st, 2014, patients with alcohol use disorder undergoing primary THA were identified and matched to a comparison group according to age, sex, and various comorbidities, resulting in 230,467 patients who were included in the study (n=38,416) and a matched-cohort (n=192,051). Outcomes of interest included comparison of LOS, 90-day medical and 2-year implant-related complications, and costs. A P-value less than 0.002 was considered statistically significant. Results Patients with alcohol use disorder had longer in-hospital LOS (4 days vs 3 days; P<0.0001) as well as higher frequency and odds ratio (OR) of 90-day medical (45.94% vs 12.25%; OR, 2.89; P<0.0001) and 2-year implant-related complications (17.71% vs 8.46%; OR, 1.97; P<0.0001). Patients in the study group incurred higher 90-day costs of care ($17,492.63 vs $14,921.88; P<0.0001). Conclusion With the growing prevalence of alcohol use disorder in the United States, the current investigation can be utilized to evaluate the need for interventions prior to THA which can potentially minimize the rates of morbidity and mortality within this population.
Collapse
Affiliation(s)
- Andrew R Horn
- Department of Orthopaedic Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| | - Keith B Diamond
- Department of Orthopaedic Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| | - Mitchell K Ng
- Department of Orthopaedic Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| | - Rushabh M Vakharia
- Department of Orthopaedic Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| | - Michael A Mont
- Department of Orthopaedic Surgery, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Orry Erez
- Department of Orthopaedic Surgery, Maimonides Medical Center, Brooklyn, NY, USA
| |
Collapse
|
23
|
Brechtel L, Poupore N, Monroe M, Knisely K, Sanders C, Edrissi C, Rathfoot C, Nathaniel TI. Role of dyslipidemia in ischemic stroke patients treated in the telestroke network. Adv Med Sci 2021; 66:254-261. [PMID: 33940526 DOI: 10.1016/j.advms.2021.04.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 03/23/2021] [Accepted: 04/20/2021] [Indexed: 01/10/2023]
Abstract
PURPOSE The relationship between the telestroke technology and clinical risk factors in a dysplipidemic ischemic stroke population and neurologic outcomes is not fully understood. This issue was investigated in this study. PATIENTS AND METHODS We analyzed retrospective data collected from a regional stroke registry to identify demographic and clinical risk factors in patients with improving (NIHSS ≤ 7) or worsening (NIHSS > 7) neurologic outcome in dyslipidemic ischemic stroke population. We used logistic multivariate models to identify independent predictors of improving or worsening outcome based on dyslipidemia disease status in ischemic stroke patients. RESULTS In the adjusted analysis for dyslipidemic ischemic stroke population, cholesterol reducer use (odd ratio; [OR] = 0.393, 95% confidence interval [CI], 0.176-0.879, P = 0.023) and direct admission (OR = 0.435, 95% CI, 0.199-0.953, P = 0.037) were more likely to be associated with neurologic improvement and no clinical or demographic factors were associated with poor neurologic outcome in dyslipidemic ischemic stroke patients treated in the telestroke network. For the ischemic stroke population without dyslipidemia, increasing age (OR = 1.070, 95% CI, 1.031-1.109, P < 0.001), coronary artery disease (OR = 3.633, 95% CI, 1.307-10.099, P = 0.013), history of drug or alcohol abuse (OR = 6.548, 95% CI, 1.106-38.777, P = 0.038), and improvement in ambulatory outcome (OR = 2.880, 95% CI, 1.183-7.010, P = 0.020) were associated with worsening neurological functions, while being Caucasian (OR = 0.294, 95% CI, 0.098-0.882, P = 0.029) was associated with improving neurologic functions. CONCLUSION Demographic and clinical risk factors among the dysplipidemic ischemic stroke population in the telestroke network were not associated with worsening neurologic functions.
Collapse
|
24
|
Lanquetin A, Leclercq S, de Timary P, Segobin S, Naveau M, Coulbault L, Maccioni P, Lorrai I, Colombo G, Vivien D, Rubio M, Pitel AL. Role of inflammation in alcohol-related brain abnormalities: a translational study. Brain Commun 2021; 3:fcab154. [PMID: 34396111 PMCID: PMC8361421 DOI: 10.1093/braincomms/fcab154] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 04/30/2021] [Accepted: 05/24/2021] [Indexed: 12/19/2022] Open
Abstract
Brain abnormalities observed in alcohol use disorder are highly heterogeneous in nature and severity, possibly because chronic alcohol consumption also affects peripheral organs leading to comorbidities that can result in exacerbated brain alterations. Despite numerous studies focussing on the effects of alcohol on the brain or liver, few studies have simultaneously examined liver function and brain damage in alcohol use disorder, and even fewer investigated the relationship between them except in hepatic encephalopathy. And yet, liver dysfunction may be a risk factor for the development of alcohol-related neuropsychological deficits and brain damage well before the development of liver cirrhosis, and potentially through inflammatory responses. The use of animal models enables a better understanding of the pathophysiological mechanisms underlying liver–brain relationships in alcohol use disorder, and more particularly of the inflammatory response at the tissue, cerebral and hepatic levels. The objective of this translational study was to investigate, both in alcohol use disorder patients and in a validated animal model of alcohol use disorder, the links between peripheral inflammation, liver damage and brain alterations. To do this, we conducted an in vivo neuroimaging examination and biological measures to evaluate brain volumes, liver fibrosis and peripheral cytokines in alcohol use disorder patients. In selectively bred Sardinian alcohol-preferring rats, we carried out ex vivo neuroimaging examination and immunohistochemistry to evaluate brain and liver inflammatory responses after chronic (50 consecutive weeks) alcohol drinking. In recently abstinent and non-cirrhotic alcohol use disorder patients, the score of liver fibrosis positively correlated with subcortical regions volumes (especially in right and left putamen) and level of circulating proinflammatory cytokines. In Sardinian alcohol-preferring rats, we found macrostructural brain damage and microstructural white matter abnormalities similar to those found in alcohol use disorder patients. In addition, in agreement with the results of peripheral inflammation observed in the patients, we revealed, in Sardinian alcohol-preferring rats, inflammatory responses in the brain and liver were caused by chronic alcohol consumption. Since the liver is the main source of cytokines in the human body, these results suggest a relationship between liver dysfunction and brain damage in alcohol use disorder patients, even in the absence of major liver disease. These findings encourage considering new therapeutic strategies aiming at treating peripheral organs to limit alcohol-related brain damage.
Collapse
Affiliation(s)
- Anastasia Lanquetin
- Normandie Univ, UNICAEN, INSERM, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France
| | - Sophie Leclercq
- Institute of Neuroscience and Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Philippe de Timary
- Institute of Neuroscience and Louvain Drug Research Institute, UCLouvain, Université Catholique de Louvain, Brussels, Belgium
| | - Shailendra Segobin
- Normandie Univ, UNICAEN, PSL Université Paris, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France
| | - Mikaël Naveau
- Normandie Univ UNICAEN, CNRS, UMS 3408, GIP Cyceron, Caen, France
| | - Laurent Coulbault
- Caen University Hospital, Biochemistry Department, Normandie University, UNICAEN, EA 4650, Caen, France
| | - Paola Maccioni
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, 09042 Monserrato, CA, Italy
| | - Irene Lorrai
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, 09042 Monserrato, CA, Italy
| | - Giancarlo Colombo
- Neuroscience Institute, Section of Cagliari, National Research Council of Italy, 09042 Monserrato, CA, Italy
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France.,Department of Clinical Research, CHU Côte de Nacre, Caen 14000, France
| | - Marina Rubio
- Normandie Univ, UNICAEN, INSERM, PhIND "Physiopathology and Imaging of Neurological Disorders", Institut Blood and Brain @ Caen-Normandie, Cyceron, 14000 Caen, France
| | - Anne-Lise Pitel
- Normandie Univ, UNICAEN, PSL Université Paris, EPHE, INSERM, U1077, CHU de Caen, GIP Cyceron, Neuropsychologie et Imagerie de la Mémoire Humaine, 14000 Caen, France.,Institut Universitaire de France (IUF), Paris 75231, France
| |
Collapse
|
25
|
Filling the gaps on stroke research: Focus on inflammation and immunity. Brain Behav Immun 2021; 91:649-667. [PMID: 33017613 PMCID: PMC7531595 DOI: 10.1016/j.bbi.2020.09.025] [Citation(s) in RCA: 73] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 09/10/2020] [Accepted: 09/23/2020] [Indexed: 02/08/2023] Open
Abstract
For the last two decades, researchers have placed hopes in a new era in which a combination of reperfusion and neuroprotection would revolutionize the treatment of stroke. Nevertheless, despite the thousands of papers available in the literature showing positive results in preclinical stroke models, randomized clinical trials have failed to show efficacy. It seems clear now that the existing data obtained in preclinical research have depicted an incomplete picture of stroke pathophysiology. In order to ameliorate bench-to-bed translation, in this review we first describe the main actors on stroke inflammatory and immune responses based on the available preclinical data, highlighting the fact that the link between leukocyte infiltration, lesion volume and neurological outcome remains unclear. We then describe what is known on neuroinflammation and immune responses in stroke patients, and summarize the results of the clinical trials on immunomodulatory drugs. In order to understand the gap between clinical trials and preclinical results on stroke, we discuss in detail the experimental results that served as the basis for the summarized clinical trials on immunomodulatory drugs, focusing on (i) experimental stroke models, (ii) the timing and selection of outcome measuring, (iii) alternative entry routes for leukocytes into the ischemic region, and (iv) factors affecting stroke outcome such as gender differences, ageing, comorbidities like hypertension and diabetes, obesity, tobacco, alcohol consumption and previous infections like Covid-19. We can do better for stroke treatment, especially when targeting inflammation following stroke. We need to re-think the design of stroke experimental setups, notably by (i) using clinically relevant models of stroke, (ii) including both radiological and neurological outcomes, (iii) performing long-term follow-up studies, (iv) conducting large-scale preclinical stroke trials, and (v) including stroke comorbidities in preclinical research.
Collapse
|