1
|
Castedo N, Alfonso A, Alvariño R, Vieytes MR, Botana LM. Cyclophilin A and C are the Main Components of Extracellular Vesicles in Response to Hyperglycemia in BV2 Microglial Cells. Mol Neurobiol 2025:10.1007/s12035-025-04921-6. [PMID: 40199808 DOI: 10.1007/s12035-025-04921-6] [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: 11/21/2024] [Accepted: 04/03/2025] [Indexed: 04/10/2025]
Abstract
Cyclophilins (Cyps) and CD147 receptor play a crucial role in the inflammatory responses. Chronic inflammation causes tissue damage and is a common condition of several inflammation-based pathologies as diabetes or Alzheimer´s disease. Under high glucose (HG) conditions, microglia is activated and releases inflammatory mediators. In this process the role of Cyps is unknown, so this study was aimed to investigate the profile of Cyps in microglia and their release through extracellular vesicles (EVs) under hyperglycemia. An increase in reactive oxygen species (ROS) and nitric oxide (NO) levels was observed when BV2 glia cells were incubated with HG concentration. These effects were mitigated by the Cyps inhibitor cyclosporine A (CsA), suggesting the implication of Cyps in BV2 activation. In these conditions the intracellular expression of CypA, B, C and D, as well as the membrane expression of CD147 receptor was increased. In addition, only CypA and CypC were detected in the extracellular medium. Then, the presence of Cyps inside EVs was explored as an alternative secretion route. Interestingly, under HG treatment, an increase in the levels of the four Cyps in EVs was observed. When neurons were treated with EVs derived from HG-treated glia cells, their viability was reduced and EVs were detected in cytosol neurons pointing to an EVs-Cyps neurotoxic effect. These findings provide novel insights into the relationship between Cyps and EVs in neuroinflammation in hyperglycemia conditions. The current results strengthen the role of Cyps in cell communication and its potential role in brain function under pathological conditions.
Collapse
Affiliation(s)
- Noelia Castedo
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Amparo Alfonso
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Rebeca Alvariño
- Departamento de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España.
| | - Mercedes R Vieytes
- Departamento de Fisiología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España
| | - Luis M Botana
- Departamento de Farmacología, Facultad de Veterinaria, IDIS, Universidad de Santiago de Compostela, Lugo, 27002, España.
| |
Collapse
|
2
|
Liu Y, Qi L, Li Z, Yong VW, Xue M. Crosstalk Between Matrix Metalloproteinases and Their Inducer EMMPRIN/CD147: a Promising Therapeutic Target for Intracerebral Hemorrhage. Transl Stroke Res 2025; 16:557-567. [PMID: 38100014 DOI: 10.1007/s12975-023-01225-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 12/05/2023] [Indexed: 04/08/2025]
Abstract
Intracerebral hemorrhage (ICH) is characterized by the disruption of cerebrovascular integrity, resulting in hematoma enlargement, edema formation, and physical damage in the brain parenchyma. Primary ICH also leads to secondary brain injury contributed by oxidative stress, dysregulated immune responses, and proteolysis. In this context, matrix metalloproteinases (MMPs) represent a ubiquitous superfamily of structurally related zinc-dependent endopeptidases capable of degrading all components of the extracellular matrix. They disrupt the blood-brain barrier and promote neuroinflammation. Importantly, several MMP members are upregulated following ICH, and members may have different functions at specific periods in ICH. Hence, the modulation and function of MMPs are more complex than expected. Extracellular matrix metalloproteinase inducer (EMMPRIN, CD147) is a transmembrane glycoprotein that induces the production of MMPs. In this review, we systematically discuss the biology and functions of MMPs and EMMPRIN/CD147 in ICH and the complex crosstalk between them.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China
| | - Lingxiao Qi
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, Alberta, Canada.
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, Zhengzhou, Henan, China.
| |
Collapse
|
3
|
Xu W, Guo Y, Zhao L, Fu R, Qin X, Zhang Y, Cheng X, Xu S. The Aging Immune System: A Critical Attack on Ischemic Stroke. Mol Neurobiol 2025; 62:3322-3342. [PMID: 39271626 DOI: 10.1007/s12035-024-04464-2] [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: 11/03/2023] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Ischemic stroke caused by cerebrovascular embolism is an age-related disease with high rates of disability and mortality. Although the mechanisms of immune and inflammatory development after stroke have been of great interest, most studies have neglected the critical and unavoidable factor of age. As the global aging trend intensifies, the number of stroke patients is constantly increasing, emphasizing the urgency of finding effective measures to address the needs of elderly stroke patients. The concept of "immunosenescence" appears to explain the worse stroke outcomes in older individuals. Immune remodeling due to aging involves dynamic changes at all levels of the immune system, and the overall consequences of central (brain-resident) and peripheral (non-brain-resident) immune cells in stroke vary according to the age of the individual. Lastly, the review outlines recent strategies aimed at immunosenescence to improve stroke prognosis.
Collapse
Affiliation(s)
- Wenzhe Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoli Qin
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
4
|
Qin Q, Feng M, Zhang K, Mo Z, Liu Y, Ma Y, Liu X. Basigin in cerebrovascular diseases: Roles, mechanisms, and therapeutic target potential. Eur J Pharmacol 2025; 989:177232. [PMID: 39734038 DOI: 10.1016/j.ejphar.2024.177232] [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: 09/22/2024] [Revised: 11/24/2024] [Accepted: 12/23/2024] [Indexed: 12/31/2024]
Abstract
Cerebrovascular diseases are major global health issues, responsible for significant morbidity and mortality. Basigin (additionally called CD147 or EMMPRIN) is a glycosylated transmembrane protein that facilitates intercellular communication. Recent research has highlighted the critical role of Basigin in inducing matrix metalloproteinases (MMPs), which contribute to the progression of cerebrovascular diseases. Consequently, Basigin has emerged as a promising therapeutic target for these conditions. However, inhibiting the pivotal role of Basigin in mediating cerebrovascular disease is an urgent area of investigation. In this review, we systematically examine the pathological mechanisms by which Basigin contributes to the development of cerebrovascular diseases. We present evidence demonstrating the protective effect of targeted inhibition of Basigin in these conditions and suggest future research directions.
Collapse
Affiliation(s)
- Qi Qin
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China
| | - Mengzhao Feng
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China
| | - Kaiyuan Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China
| | - Zhizhun Mo
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Shenzhen City, Guangdong Province, 518033, China
| | - Yuxiang Liu
- Emergency Department, Shenzhen Traditional Chinese Medicine Hospital, The fourth Clinical Medical College of Guangzhou University of Chinese Medicine, No.1 Fuhua Road, Shenzhen City, Guangdong Province, 518033, China
| | - Yinzhong Ma
- Institute of Medicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen City, Guangdong Province, 518055, China.
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, No.1 Jianshe East Road, Zhengzhou City, Henan Province, 450000, China.
| |
Collapse
|
5
|
Mora VP, Kalergis AM, Bohmwald K. Neurological Impact of Respiratory Viruses: Insights into Glial Cell Responses in the Central Nervous System. Microorganisms 2024; 12:1713. [PMID: 39203555 PMCID: PMC11356956 DOI: 10.3390/microorganisms12081713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 09/03/2024] Open
Abstract
Respiratory viral infections pose a significant public health threat, particularly in children and older adults, with high mortality rates. Some of these pathogens are the human respiratory syncytial virus (hRSV), severe acute respiratory coronavirus-2 (SARS-CoV-2), influenza viruses (IV), human parvovirus B19 (B19V), and human bocavirus 1 (HBoV1). These viruses cause various respiratory symptoms, including cough, fever, bronchiolitis, and pneumonia. Notably, these viruses can also impact the central nervous system (CNS), leading to acute manifestations such as seizures, encephalopathies, encephalitis, neurological sequelae, and long-term complications. The precise mechanisms by which these viruses affect the CNS are not fully understood. Glial cells, specifically microglia and astrocytes within the CNS, play pivotal roles in maintaining brain homeostasis and regulating immune responses. Exploring how these cells interact with viral pathogens, such as hRSV, SARS-CoV-2, IVs, B19V, and HBoV1, offers crucial insights into the significant impact of respiratory viruses on the CNS. This review article examines hRSV, SARS-CoV-2, IV, B19V, and HBoV1 interactions with microglia and astrocytes, shedding light on potential neurological consequences.
Collapse
Affiliation(s)
- Valentina P. Mora
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| | - Alexis M. Kalergis
- Millennium Institute on Immunology and Immunotherapy (MIII), Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile;
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Karen Bohmwald
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago 8910060, Chile;
| |
Collapse
|
6
|
Wang Z, Zhang X, Zhang G, Zheng YJ, Zhao A, Jiang X, Gan J. Astrocyte modulation in cerebral ischemia-reperfusion injury: A promising therapeutic strategy. Exp Neurol 2024; 378:114814. [PMID: 38762094 DOI: 10.1016/j.expneurol.2024.114814] [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: 01/25/2024] [Revised: 04/03/2024] [Accepted: 05/12/2024] [Indexed: 05/20/2024]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) poses significant challenges for drug development due to its complex pathogenesis. Astrocyte involvement in CIRI pathogenesis has led to the development of novel astrocyte-targeting drug strategies. To comprehensively review the current literature, we conducted a thorough analysis from January 2012 to December 2023, identifying 82 drugs aimed at preventing and treating CIRI. These drugs target astrocytes to exert potential benefits in CIRI, and their primary actions include modulation of relevant signaling pathways to inhibit neuroinflammation and oxidative stress, reduce cerebral edema, restore blood-brain barrier integrity, suppress excitotoxicity, and regulate autophagy. Notably, active components from traditional Chinese medicines (TCM) such as Salvia miltiorrhiza, Ginkgo, and Ginseng exhibit these important pharmacological properties and show promise in the treatment of CIRI. This review highlights the potential of astrocyte-targeted drugs to ameliorate CIRI and categorizes them based on their mechanisms of action, underscoring their therapeutic potential in targeting astrocytes.
Collapse
Affiliation(s)
- Ziyu Wang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaolu Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Guangming Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yu Jia Zheng
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Anliu Zhao
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xijuan Jiang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| | - Jiali Gan
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China.
| |
Collapse
|
7
|
Hirayama Y, Le HPN, Hashimoto H, Ishii I, Koizumi S, Anzai N. Preconditioning-Induced Facilitation of Lactate Release from Astrocytes Is Essential for Brain Ischemic Tolerance. eNeuro 2024; 11:ENEURO.0494-23.2024. [PMID: 38604775 PMCID: PMC11064122 DOI: 10.1523/eneuro.0494-23.2024] [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: 11/26/2023] [Revised: 03/27/2024] [Accepted: 04/05/2024] [Indexed: 04/13/2024] Open
Abstract
A sublethal ischemic episode [termed preconditioning (PC)] protects neurons in the brain against a subsequent severe ischemic injury. This phenomenon is known as brain ischemic tolerance and has received much attention from researchers because of its robust neuroprotective effects. We have previously reported that PC activates astrocytes and subsequently upregulates P2X7 receptors, thereby leading to ischemic tolerance. However, the downstream signals of P2X7 receptors that are responsible for PC-induced ischemic tolerance remain unknown. Here, we show that PC-induced P2X7 receptor-mediated lactate release from astrocytes has an indispensable role in this event. Using a transient focal cerebral ischemia model caused by middle cerebral artery occlusion, extracellular lactate levels during severe ischemia were significantly increased in mice who experienced PC; this increase was dependent on P2X7 receptors. In addition, the intracerebroventricular injection of lactate protected against cerebral ischemic injury. In in vitro experiments, although stimulation of astrocytes with the P2X7 receptor agonist BzATP had no effect on the protein levels of monocarboxylate transporter (MCT) 1 and MCT4 (which are responsible for lactate release from astrocytes), BzATP induced the plasma membrane translocation of these MCTs via their chaperone CD147. Importantly, CD147 was increased in activated astrocytes after PC, and CD147-blocking antibody abolished the PC-induced facilitation of astrocytic lactate release and ischemic tolerance. Taken together, our findings suggest that astrocytes induce ischemic tolerance via P2X7 receptor-mediated lactate release.
Collapse
Affiliation(s)
- Yuri Hirayama
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Ha Pham Ngoc Le
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Hirofumi Hashimoto
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| | - Itsuko Ishii
- Division of Pharmacy, Chiba University Hospital, Chiba 260-8677, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan
- GLIA Center, University of Yamanashi, Yamanashi 409-3898, Japan
| | - Naohiko Anzai
- Department of Pharmacology, Chiba University Graduate School of Medicine, Chiba 260-8670, Japan
| |
Collapse
|
8
|
Li Y, Chen J, Quan X, Chen Y, Han Y, Chen J, Yang L, Xu Y, Shen X, Wang R, Zhao Y. Extracellular Vesicles Maintain Blood-Brain Barrier Integrity by the Suppression of Caveolin-1/CD147/VEGFR2/MMP Pathway After Ischemic Stroke. Int J Nanomedicine 2024; 19:1451-1467. [PMID: 38371456 PMCID: PMC10874237 DOI: 10.2147/ijn.s444009] [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: 10/10/2023] [Accepted: 02/08/2024] [Indexed: 02/20/2024] Open
Abstract
Background Ischemic stroke (IS) causes tragic death and disability worldwide. However, effective therapeutic interventions are finite. After IS, blood-brain barrier (BBB) integrity is disrupted, resulting in deteriorating neurological function. As a novel therapeutic, extracellular vesicles (EVs) have shown ideal restorative effects on BBB integrity post-stroke; however, the definite mechanisms remain ambiguous. In the present study, we investigated the curative effects and the mechanisms of EVs derived from bone marrow mesenchymal stem cells and brain endothelial cells (BMSC-EVs and BEC-EVs) on BBB integrity after acute IS. Methods EVs were isolated from BMSCs and BECs, and we investigated the therapeutic effect in vitro oxygen-glucose deprivation (OGD) insulted BECs model and in vivo rat middle cerebral artery occlusion (MCAo) model. The cell monolayer leakage, tight junction expression, and metalloproteinase (MMP) activity were evaluated, and rat brain infarct volume and neurological function were also analyzed. Results The administration of two kinds of EVs not only enhanced ZO-1 and Occludin expressions but also reduced the permeability and the activity of MMP-2/9 in OGD-insulted BECs. The amelioration of the cerebral infarction, BBB leakage, neurological function deficits, and the increasing ZO-1 and Occludin levels, as well as MMP activity inhibition was observed in MCAo rats. Additionally, the increased levels of Caveolin-1, CD147, vascular endothelial growth factor receptor 2 (VEGFR2), and vascular endothelial growth factor A (VEGFA) in isolated brain microvessels were downregulated after EVs treatment. In vitro, the employment of Caveolin-1 and CD147 siRNA partly suppressed the expressions of VEGFR2, VEGFA and MMP-2/9 activity and reduced the leakage of OGD insulted BECs and enhanced ZO-1 and Occludin expressions. Conclusion Our study firstly demonstrates that BEC and BMSC-EVs administrations maintain BBB integrity via the suppression of Caveolin-1/CD147/VEGFR2/MMP pathway after IS, and the efficacy of BMSC-EVs is superior to that of BEC-EVs.
Collapse
Affiliation(s)
- Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Jiali Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Ying Chen
- School of Health Economics and Management, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, People’s Republic of China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Jinfen Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Li Yang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Youhua Xu
- Faculty of Chinese Medicine, Macau University of Science and Technology, Taipa, Macao SAR, People’s Republic of China
| | - Xu Shen
- Jiangsu Key Laboratory of Drug Target and Drug for Degenerative Diseases, Nanjing University of Chinese Medicine, Nanjing, People’s Republic of China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People’s Republic of China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, People’s Republic of China
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, People’s Republic of China
| |
Collapse
|
9
|
Goncharov NV, Popova PI, Kudryavtsev IV, Golovkin AS, Savitskaya IV, Avdonin PP, Korf EA, Voitenko NG, Belinskaia DA, Serebryakova MK, Matveeva NV, Gerlakh NO, Anikievich NE, Gubatenko MA, Dobrylko IA, Trulioff AS, Aquino AD, Jenkins RO, Avdonin PV. Immunological Profile and Markers of Endothelial Dysfunction in Elderly Patients with Cognitive Impairments. Int J Mol Sci 2024; 25:1888. [PMID: 38339164 PMCID: PMC10855959 DOI: 10.3390/ijms25031888] [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: 12/10/2023] [Revised: 01/19/2024] [Accepted: 01/31/2024] [Indexed: 02/12/2024] Open
Abstract
The process of aging is accompanied by a dynamic restructuring of the immune response, a phenomenon known as immunosenescence. Further, damage to the endothelium can be both a cause and a consequence of many diseases, especially in elderly people. The purpose of this study was to carry out immunological and biochemical profiling of elderly people with acute ischemic stroke (AIS), chronic cerebral circulation insufficiency (CCCI), prediabetes or newly diagnosed type II diabetes mellitus (DM), and subcortical ischemic vascular dementia (SIVD). Socio-demographic, lifestyle, and cognitive data were obtained. Biochemical, hematological, and immunological analyses were carried out, and extracellular vesicles (EVs) with endothelial CD markers were assessed. The greatest number of significant deviations from conditionally healthy donors (HDs) of the same age were registered in the SIVD group, a total of 20, of which 12 were specific and six were non-specific but with maximal differences (as compared to the other three groups) from the HDs group. The non-specific deviations were for the MOCA (Montreal Cognitive Impairment Scale), the MMSE (Mini Mental State Examination) and life satisfaction self-assessment scores, a decrease of albumin levels, and ADAMTS13 (a Disintegrin and Metalloproteinase with a Thrombospondin Type 1 motif, member 13) activity, and an increase of the VWF (von Willebrand factor) level. Considering the significant changes in immunological parameters (mostly Th17-like cells) and endothelial CD markers (CD144 and CD34), vascular repair was impaired to the greatest extent in the DM group. The AIS patients showed 12 significant deviations from the HD controls, including three specific to this group. These were high NEFAs (non-esterified fatty acids) and CD31 and CD147 markers of EVs. The lowest number of deviations were registered in the CCCI group, nine in total. There were significant changes from the HD controls with no specifics to this group, and just one non-specific with a maximal difference from the control parameters, which was α1-AGP (alpha 1 acid glycoprotein, orosomucoid). Besides the DM patients, impairments of vascular repair were also registered in the CCCI and AIS patients, with a complete absence of such in patients with dementia (SIVD group). On the other hand, microvascular damage seemed to be maximal in the latter group, considering the biochemical indicators VWF and ADAMTS13. In the DM patients, a maximum immune response was registered, mainly with Th17-like cells. In the CCCI group, the reaction was not as pronounced compared to other groups of patients, which may indicate the initial stages and/or compensatory nature of organic changes (remodeling). At the same time, immunological and biochemical deviations in SIVD patients indicated a persistent remodeling in microvessels, chronic inflammation, and a significant decrease in the anabolic function of the liver and other tissues. The data obtained support two interrelated assumptions. Taking into account the primary biochemical factors that trigger the pathological processes associated with vascular pathology and related diseases, the first assumption is that purine degradation in skeletal muscle may be a major factor in the production of uric acid, followed by its production by non-muscle cells, the main of which are endothelial cells. Another assumption is that therapeutic factors that increase the levels of endothelial progenitor cells may have a therapeutic effect in reducing the risk of cerebrovascular disease and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Nikolay V. Goncharov
- Research Institute of Hygiene, Occupational Pathology and Human Ecology of the Federal Medical Biological Agency, bld 93 Kuzmolovsky, Leningrad Region 188663, Russia
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg 194223, Russia
| | | | | | | | | | - Piotr P. Avdonin
- Koltsov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow 119334, Russia
| | - Ekaterina A. Korf
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg 194223, Russia
| | - Natalia G. Voitenko
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg 194223, Russia
| | - Daria A. Belinskaia
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg 194223, Russia
| | | | | | | | | | | | - Irina A. Dobrylko
- Sechenov Institute of Evolutionary Physiology and Biochemistry of the Russian Academy of Sciences, St. Petersburg 194223, Russia
| | | | - Arthur D. Aquino
- Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Richard O. Jenkins
- School of Allied Health Sciences, De Montfort University, The Gateway, Leicester LE1 9BH, UK
| | - Pavel V. Avdonin
- Koltsov Institute of Developmental Biology of the Russian Academy of Sciences, Moscow 119334, Russia
| |
Collapse
|
10
|
Li F, Wang J, Yan YQ, Bai CZ, Guo JQ. CD147 promotes breast cancer migration and invasion by inducing epithelial-mesenchymal transition via the MAPK/ERK signaling pathway. BMC Cancer 2023; 23:1214. [PMID: 38066486 PMCID: PMC10709944 DOI: 10.1186/s12885-023-11724-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 12/06/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND CD147, a transmembrane glycoprotein, has been implicated in various cancer-related processes but its role in breast cancer remains poorly understood. Herein, we investigated the expression of CD147 in different breast cancer cell lines and explored its functional roles, including migration, invasion, drug resistance and modulation of key proteins associated with cancer progression. METHODS The expression of CD147 was assessed in MCF-10 A, BT549, MDA-MB-231 and MCF-7 breast cancer cell lines using qRT-PCR and Western blotting, following which lyposome transfections were performed, leading overexpression of CD147 in BT549 cells and knockdown of CD147 in MCF-7 cells. Scratch assays and Transwell invasion and were performed to evaluate the cells' migration and invasion abilities. Sensitivity to 5-FU was determined via CCK-8 assays, and the expression of Snail1, E-cadherin, Vimentin, MMP-9 and the MAPK/ERK pathway were analyzed by qRT-PCR and Western blotting. RESULTS Compared with normal beast epithelial cells, CD147 was highly expressed in all breast cancer cell lines, with the highest overexpression observed in MCF-7 cells and the lowest overexpression observed in BT549 cells. Overexpression of CD147 in BT549 cells increased, migration, invasion, viability and resistance to 5-FU of BT549 cells, while CD147 knockdown in MCF-7 cells reduced these properties of MCF-7 cells. Furthermore, CD147 influenced the expression of Snail1, Vimentin, E-cadherin, and MMP-9, suggesting its involvement in epithelial-mesenchymal transition (EMT) regulation. The MAPK/ERK pathway was activated by CD147 in BT549 cells, as indicated by increased p-MEK/MEK ratio and p-ERK/ERK ratio. In contrast, CD147 silencing in MCF-7 cells resulted in reduced p-MEK/MEK ratio and p-ERK/ERK ratio. CONCLUSION In summary, our findings suggest CD147 as a potential therapeutic target in breast cancer treatment, particularly in cases where drug resistance and metastasis are concerns, worthy of further explorations.
Collapse
Affiliation(s)
- Fang Li
- Department of Scientific Research, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, PR China.
| | - Jing Wang
- Department of Neurosurgery, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, PR China
| | - Yu-Qiong Yan
- Key Laboratory of Chemical Biology and Molecular Engineering of Ministry of Education, Institute of Biotechnology, Shanxi University, Taiyuan, 030006, PR China
| | - Chong-Zhi Bai
- Central Laboratory, Shanxi Province Hospital of Traditional Chinese Medicine, Taiyuan, 030012, PR China
| | - Ji-Qiang Guo
- Department of Clinical Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, 030032, PR China
| |
Collapse
|
11
|
Yao C, Liu X, Tang Y, Wang C, Duan C, Liu X, Chen M, Zhou Y, Tang E, Xiang Y, Li Y, Ji A, Cai T. Lipopolysaccharide induces inflammatory microglial activation through CD147-mediated matrix metalloproteinase expression. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:35352-35365. [PMID: 36534246 PMCID: PMC9761036 DOI: 10.1007/s11356-022-24292-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 11/14/2022] [Indexed: 06/17/2023]
Abstract
Microglia-mediated neuroinflammation plays a vital role in the pathophysiological processes of multiple neurodegenerative diseases. Lipopolysaccharide (LPS) is an environmental poison that can induce inflammatory microglial activation. Matrix metalloproteinases (MMPs) are vital factors regulating microglial activation, and CD147 is a key MMP inducer, which can induce inflammation by inducing MMPs. However, whether it is involved in the regulation of microglial activation has not been reported. In this study, the role of CD147 in LPS-induced microglial inflammatory activation was investigated by establishing in vivo and in vitro models. The results suggested that LPS-induced microglial activation was accompanied by the induction of CD147 expression while the inhibition of CD147 expression could inhibit LPS-induced microglial inflammatory activation. In addition, the results also indicated that the role of CD147 in LPS-induced pro-inflammatory activation of microglia was related to its downstream MMP-3, MMP-8, and autophagy. Furthermore, the inhibition of MMP-3, MMP-8, and autophagy attenuated LPS-induced inflammatory activation of microglia. At the same time, there was a certain interaction between MMPs and autophagy, which is shown that inhibiting the expression of MMPs could inhibit autophagy, whereas inhibiting autophagy could inhibit the expression of MMPs. Taken together, we provided the first evidence that CD147/MMPs can be involved in LPS-induced inflammatory activation of microglia through an autophagy-dependent manner.
Collapse
Affiliation(s)
- Chunyan Yao
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Xiaoling Liu
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Yan Tang
- Experimental Teaching Center, School of Public Health, Southwest Medical University, Luzhou, China
| | - Chunmei Wang
- Experimental Teaching Center, School of Public Health, Southwest Medical University, Luzhou, China
| | - Chenggang Duan
- Department of Pathophysiology, Southwest Medical University, Luzhou, China
| | - Xiaoyan Liu
- The Research Center for Preclinical Medicine, Southwest Medical University, Luzhou, China
| | - Mingliang Chen
- Department of Chemical Defense Medicine, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Yumeng Zhou
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Enjie Tang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Ying Xiang
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Yafei Li
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China
| | - Ailing Ji
- Department of Preventive Medicine & Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Tongjian Cai
- Department of Epidemiology, College of Preventive Medicine, Army Medical University, Third Military Medical University), Chongqing, China.
| |
Collapse
|
12
|
Zhang W, Wu Q, Hao S, Chen S. The hallmark and crosstalk of immune cells after intracerebral hemorrhage: Immunotherapy perspectives. Front Neurosci 2023; 16:1117999. [PMID: 36711145 PMCID: PMC9877537 DOI: 10.3389/fnins.2022.1117999] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 12/30/2022] [Indexed: 01/13/2023] Open
Abstract
Intracerebral hemorrhage (ICH) is one of the most dangerous types of strokes with a high morbidity and mortality rate. Currently, the treatment of ICH is not well developed, mainly because its mechanisms are still unclear. Inflammation is one of the main types of secondary injury after ICH and catalyzes the adverse consequences of ICH. A large number of immune cells are involved in neuroinflammation, such as microglia, astrocytes, oligodendrocytes, lymphocytes, macrophages, and neutrophils. Nevertheless, the characteristics and crosstalk of immune cells have not been fully elucidated. In this review, we endeavor to delve into the respective characteristics of immune cells and their interactions in neuroimmune inflammation, and further elucidate favorable immunotherapeutic approaches regarding ICH, and finally present an outlook.
Collapse
Affiliation(s)
- Wenqing Zhang
- School of Medicine, Chongqing University, Chongqing, China,Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Qingyuan Wu
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China
| | - Shilei Hao
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, China,*Correspondence: Shilei Hao,
| | - Shengli Chen
- Department of Neurology, Chongqing University Three Gorges Hospital, Chongqing, China,Shengli Chen,
| |
Collapse
|
13
|
Asgari R, Vaisi-Raygani A, Aleagha MSE, Mohammadi P, Bakhtiari M, Arghiani N. CD147 and MMPs as key factors in physiological and pathological processes. Biomed Pharmacother 2023; 157:113983. [PMID: 36370522 DOI: 10.1016/j.biopha.2022.113983] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 11/03/2022] [Accepted: 11/04/2022] [Indexed: 11/11/2022] Open
Abstract
Cluster of differentiation 147 (CD147) or extracellular matrix metalloproteinase inducer (EMMPRIN) is a transmembrane glycoprotein that induces the synthesis of matrix metalloproteinases (MMPs). MMPs, as zinc-dependent proteases and versatile enzymes, play critical roles in the degradation of the extracellular matrix (ECM) components, cleaving of the receptors of cellular surfaces, signaling molecules, and other precursor proteins, which may lead to attenuation or activation of such targets. CD147 and MMPs play essential roles in physiological and pathological conditions and any disorder in the expression, synthesis, or function of CD147 and MMPs may be associated with various types of disease. In this review, we have focused on the roles of CD147 and MMPs in some major physiological and pathological processes.
Collapse
Affiliation(s)
- Rezvan Asgari
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Asad Vaisi-Raygani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mohammad Sajad Emami Aleagha
- Department of Clinical Biochemistry, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Pantea Mohammadi
- Medical Biology Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Mitra Bakhtiari
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| | - Nahid Arghiani
- Department of Molecular Biosciences, the Wenner-Gren Institute, Stockholm University, Stockholm, Sweden; School of Life Science, Department of Biochemistry and Biomedicine, University of Sussex, Brighton, United Kingdom.
| |
Collapse
|
14
|
Liu Y, Mu Y, Li Z, Yong VW, Xue M. Extracellular matrix metalloproteinase inducer in brain ischemia and intracerebral hemorrhage. Front Immunol 2022; 13:986469. [PMID: 36119117 PMCID: PMC9471314 DOI: 10.3389/fimmu.2022.986469] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 08/11/2022] [Indexed: 12/13/2022] Open
Abstract
Increasing evidence from preclinical and clinical studies link neuroinflammation to secondary brain injury after stroke, which includes brain ischemia and intracerebral hemorrhage (ICH). Extracellular matrix metalloproteinase inducer (EMMPRIN), a cell surface transmembrane protein, is a key factor in neuroinflammation. It is widely elevated in several cell types after stroke. The increased EMMPRIN appears to regulate the expression of matrix metalloproteinases (MMPs) and exacerbate the pathology of stroke-induced blood-brain barrier dysfunction, microvascular thrombosis and neuroinflammation. In light of the neurological effects of EMMPRIN, we present in this review the complex network of roles that EMMPRIN has in brain ischemia and ICH. We first introduce the structural features and biological roles of EMMPRIN, followed by a description of the increased expression of EMMPRIN in brain ischemia and ICH. Next, we discuss the pathophysiological roles of EMMPRIN in brain ischemia and ICH. In addition, we summarize several important treatments for stroke that target the EMMPRIN signaling pathway. Finally, we suggest that EMMPRIN may have prospects as a biomarker of stroke injury. Overall, this review collates experimental and clinical evidence of the role of EMMPRIN in stroke and provides insights into its pathological mechanisms.
Collapse
Affiliation(s)
- Yang Liu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanling Mu
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhe Li
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Voon Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada
- *Correspondence: Voon Wee Yong, ; Mengzhou Xue,
| | - Mengzhou Xue
- Department of Cerebrovascular Diseases, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Henan International Joint Laboratory of Intracerebral Hemorrhage and Brain Injury, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Voon Wee Yong, ; Mengzhou Xue,
| |
Collapse
|
15
|
Bianchi L, Altera A, Barone V, Bonente D, Bacci T, De Benedetto E, Bini L, Tosi GM, Galvagni F, Bertelli E. Untangling the Extracellular Matrix of Idiopathic Epiretinal Membrane: A Path Winding among Structure, Interactomics and Translational Medicine. Cells 2022; 11:cells11162531. [PMID: 36010606 PMCID: PMC9406781 DOI: 10.3390/cells11162531] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 08/10/2022] [Accepted: 08/12/2022] [Indexed: 12/16/2022] Open
Abstract
Idiopathic epiretinal membranes (iERMs) are fibrocellular sheets of tissue that develop at the vitreoretinal interface. The iERMs consist of cells and an extracellular matrix (ECM) formed by a complex array of structural proteins and a large number of proteins that regulate cell–matrix interaction, matrix deposition and remodelling. Many components of the ECM tend to produce a layered pattern that can influence the tractional properties of the membranes. We applied a bioinformatics approach on a list of proteins previously identified with an MS-based proteomic analysis on samples of iERM to report the interactome of some key proteins. The performed pathway analysis highlights interactions occurring among ECM molecules, their cell receptors and intra- or extracellular proteins that may play a role in matrix biology in this special context. In particular, integrin β1, cathepsin B, epidermal growth factor receptor, protein-glutamine gamma-glutamyltransferase 2 and prolow-density lipoprotein receptor-related protein 1 are key hubs in the outlined protein–protein cross-talks. A section on the biomarkers that can be found in the vitreous humor of patients affected by iERM and that can modulate matrix deposition is also presented. Finally, translational medicine in iERM treatment has been summed up taking stock of the techniques that have been proposed for pharmacologic vitreolysis.
Collapse
Affiliation(s)
- Laura Bianchi
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Annalisa Altera
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Virginia Barone
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Denise Bonente
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
| | - Tommaso Bacci
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Elena De Benedetto
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Luca Bini
- Department of Life Sciences, University of Siena, 53100 Siena, Italy
| | - Gian Marco Tosi
- Department of Medicine, Surgery and Neuroscience, University of Siena, 53100 Siena, Italy
| | - Federico Galvagni
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, 53100 Siena, Italy
| | - Eugenio Bertelli
- Department of Molecular and Developmental Medicine, University of Siena, 53100 Siena, Italy
- Correspondence:
| |
Collapse
|
16
|
Neuroprotective Effects of Chlorogenic Acid in a Mouse Model of Intracerebral Hemorrhage Associated with Reduced Extracellular Matrix Metalloproteinase Inducer. Biomolecules 2022; 12:biom12081020. [PMID: 35892330 PMCID: PMC9332591 DOI: 10.3390/biom12081020] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
Chlorogenic acid (CGA) has been reported to have various biological activities, such as anti-inflammatory, anti-oxidant and anti-apoptosis effects. However, the role of CGA in intracerebral hemorrhage (ICH) and the underlying mechanisms remain undiscovered. The current study aims to investigate the effect of CGA on neuroinflammation and neuronal apoptosis after inhibition of EMMPRIN in a collagenase-induced ICH mouse model. Dose optimization data showed that intraperitoneal administration of CGA (30 mg/kg) significantly attenuated neurological impairments and reduced brain water content at 24 h and 72 h compared with ICH mice given vehicle. Western blot and immunofluorescence analyses revealed that CGA remarkably decreased the expression of extracellular matrix metalloproteinase inducer (EMMPRIN) in perihematomal areas at 72 h after ICH. CGA also reduced the expression of matrix metalloproteinases-2/9 (MMP-2/9) at 72 h after ICH. CGA diminished Evans blue dye extravasation and reduced the loss of zonula occludens-1 (ZO-1) and occludin. CGA-treated mice had fewer activated Iba-1-positive microglia and MPO-positive neutrophils. Finally, CGA suppressed cell death around the hematoma and reduced overall brain injury. These outcomes highlight that CGA treatment confers neuroprotection in ICH likely by inhibiting expression of EMMPRIN and MMP-2/9, and alleviating neuroinflammation, blood–brain barrier (BBB) disruption, cell death and brain injury.
Collapse
|
17
|
DeLong JH, Ohashi SN, O'Connor KC, Sansing LH. Inflammatory Responses After Ischemic Stroke. Semin Immunopathol 2022; 44:625-648. [PMID: 35767089 DOI: 10.1007/s00281-022-00943-7] [Citation(s) in RCA: 85] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 04/20/2022] [Indexed: 12/25/2022]
Abstract
Ischemic stroke generates an immune response that contributes to neuronal loss as well as tissue repair. This is a complex process involving a range of cell types and effector molecules and impacts tissues outside of the CNS. Recent reviews address specific aspects of this response, but several years have passed and important advances have been made since a high-level review has summarized the overall state of the field. The present review examines the initiation of the inflammatory response after ischemic stroke, the complex impacts of leukocytes on patient outcome, and the potential of basic science discoveries to impact the development of therapeutics. The information summarized here is derived from broad PubMed searches and aims to reflect recent research advances in an unbiased manner. We highlight valuable recent discoveries and identify gaps in knowledge that have the potential to advance our understanding of this disease and therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Jonathan Howard DeLong
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Sarah Naomi Ohashi
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kevin Charles O'Connor
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lauren Hachmann Sansing
- Departments of Neurology and Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
18
|
Badeti S, Jiang Q, Naghizadeh A, Tseng HC, Bushkin Y, Marras SAE, Nisa A, Tyagi S, Chen F, Romanienko P, Yehia G, Evans D, Lopez-Gonzalez M, Alland D, Russo R, Gause W, Shi L, Liu D. Development of a novel human CD147 knock-in NSG mouse model to test SARS-CoV-2 viral infection. Cell Biosci 2022; 12:88. [PMID: 35690792 PMCID: PMC9187929 DOI: 10.1186/s13578-022-00822-6] [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: 03/15/2022] [Accepted: 05/27/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND An animal model that can mimic the SARS-CoV-2 infection in humans is critical to understanding the rapidly evolving SARS-CoV-2 virus and for development of prophylactic and therapeutic strategies to combat emerging mutants. Studies show that the spike proteins of SARS-CoV and SARS-CoV-2 bind to human angiotensin-converting enzyme 2 (hACE2, a well-recognized, functional receptor for SARS-CoV and SARS-CoV-2) to mediate viral entry. Several hACE2 transgenic (hACE2Tg) mouse models are being widely used, which are clearly invaluable. However, the hACE2Tg mouse model cannot fully explain: (1) low expression of ACE2 observed in human lung and heart, but lung or heart failure occurs frequently in severe COVID-19 patients; (2) low expression of ACE2 on immune cells, but lymphocytopenia occurs frequently in COVID-19 patients; and (3) hACE2Tg mice do not mimic the natural course of SARS-CoV-2 infection in humans. Moreover, one of most outstanding features of coronavirus infection is the diversity of receptor usage, which includes the newly proposed human CD147 (hCD147) as a possible co-receptor for SARS-CoV-2 entry. It is still debatable whether CD147 can serve as a functional receptor for SARS-CoV-2 infection or entry. RESULTS Here we successfully generated a hCD147 knock-in mouse model (hCD147KI) in the NOD-scid IL2Rgammanull (NSG) background. In this hCD147KI-NSG mouse model, the hCD147 genetic sequence was placed downstream of the endogenous mouse promoter for mouse CD147 (mCD147), which creates an in vivo model that may better recapitulate physiological expression of hCD147 proteins at the molecular level compared to the existing and well-studied K18-hACE2-B6 (JAX) model. In addition, the hCD147KI-NSG mouse model allows further study of SARS-CoV-2 in the immunodeficiency condition which may assist our understanding of this virus in the context of high-risk populations in immunosuppressed states. Our data show (1) the human CD147 protein is expressed in various organs (including bronchiolar epithelial cells) in hCD147KI-NSG mice by immunohistochemical staining and flow cytometry; (2) hCD147KI-NSG mice are marginally sensitive to SARS-CoV-2 infection compared to WT-NSG littermates characterized by increased viral copies by qRT-PCR and moderate body weight decline compared to baseline; (3) a significant increase in leukocytes in the lungs of hCD147KI-NSG mice, compared to infected WT-NSG mice. CONCLUSIONS hCD147KI-NSG mice are more sensitive to COVID-19 infection compared to WT-NSG mice. The hCD147KI-NSG mouse model can serve as an additional animal model for further interrogation whether CD147 serve as an independent functional receptor or accessory receptor for SARS-CoV-2 entry and immune responses.
Collapse
Affiliation(s)
- Saiaditya Badeti
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA
- School of Graduate Studies, Biomedical and Health Sciences, Rutgers University, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Qingkui Jiang
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Alireza Naghizadeh
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Hsiang-Chi Tseng
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Yuri Bushkin
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Salvatore A E Marras
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Annuurun Nisa
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Sanjay Tyagi
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Fei Chen
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Peter Romanienko
- Genome Editing Shared Resources, Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Ghassan Yehia
- Genome Editing Shared Resources, Cancer Institute of New Jersey, Rutgers University, New Brunswick, NJ, 08901, USA
| | - Deborah Evans
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Moises Lopez-Gonzalez
- Regional Bio-Containment Laboratory, Center for COVID-19 Response and Pandemic Preparedness (CCRP2), Rutgers-New Jersey Medical School, Newark, NJ, 07103, USA
| | - David Alland
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Riccardo Russo
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - William Gause
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA
| | - Lanbo Shi
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA.
| | - Dongfang Liu
- Department of Pathology, Immunology and Laboratory Medicine, Center for Immunity and Inflammation, Rutgers-New Jersey Medical School, Rutgers University, 205 S. Orange Ave., CC-H1218, Newark, NJ, 07103, USA.
- School of Graduate Studies, Biomedical and Health Sciences, Rutgers University, Newark, NJ, 07103, USA.
- Center for Immunity and Inflammation, New Jersey Medical School, Rutgers University, Newark, NJ, 07103, USA.
| |
Collapse
|
19
|
Liu Y, Bai Q, Yong VW, Xue M. EMMPRIN Promotes the Expression of MMP-9 and Exacerbates Neurological Dysfunction in a Mouse Model of Intracerebral Hemorrhage. Neurochem Res 2022; 47:2383-2395. [PMID: 35608790 DOI: 10.1007/s11064-022-03630-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 04/02/2022] [Accepted: 05/07/2022] [Indexed: 12/23/2022]
Abstract
Extracellular matrix metalloproteinase inducer (EMMPRIN) has been shown to be a vital inflammatory mediator in several neurological and neurodegenerative diseases. However, the role of EMMPRIN in intracerebral hemorrhage (ICH) remains unexplored. In this study, we aimed to exploit a highly selective monoclonal anti-EMMPRIN antibody to functionally inhibit EMMPRIN activity and thus that of MMPs as the downstream effector. To induce ICH pathology, adult C57BL/6 male mice were injected with collagenase type VII or saline as control into the right basal ganglia and were euthanized at different time points. The anti-EMMPRIN monoclonal antibody was intravenously injected once daily for 3 days to block the expression of EMMPRIN initiating at 4 h post-ICH. Western blot and immunofluorescence analysis results revealed that EMMPRIN expression was significantly increased surrounding the hematoma at 3 and 7 d time points after ICH when compared to the saline treated control group. EMMPRIN expression was co-localized with GFAP (astrocytes) and Iba1 (microglia) at 3 d time point post-ICH, but not in the control group mice. The co-localization of EMMPRIN with CD31 in endothelial cells occurred in both groups and was higher in the ICH brain. However, EMMPRIN expression was not detected in neurons from either group. The inhibition of EMMPRIN reduced the expression of MMP-9, the number of infiltrated neutrophils, the degree of brain injury and promoted neurological recovery after ICH. In conclusion, EMMPRIN could mediate the upregulation of MMP-9 and exacerbate neurological dysfunction in a mouse model of experimental ICH. Furthermore, blocking EMMPRIN reduced brain injury and subsequently promoted neurological recovery in ICH mice brains. These outcomes highlight that inhibition of EMMPRIN can be a potential therapeutic intervention strategy to regulate MMP-9's pathological roles during ICH.
Collapse
Affiliation(s)
- Yang Liu
- Departments of Cerebrovascular Diseases and Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China.,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - Qian Bai
- Departments of Cerebrovascular Diseases and Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China.,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China
| | - V Wee Yong
- Hotchkiss Brain Institute and Department of Clinical Neurosciences, University of Calgary, Calgary, AB, Canada.
| | - Mengzhou Xue
- Departments of Cerebrovascular Diseases and Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, 2 Jingba Road, Zhengzhou, 450001, Henan, China. .,Henan Medical Key Laboratory of Translational Cerebrovascular Diseases, Zhengzhou, Henan, China.
| |
Collapse
|
20
|
Badeti S, Jiang Q, Naghizadeh A, Tseng HC, Bushkin Y, Marras SAE, Nisa A, Tyagi S, Chen F, Romanienko P, Yehia G, Evans D, Lopez-Gonzalez M, Alland D, Russo R, Gause W, Shi L, Liu D. Development of a Novel Human CD147 Knock-in NSG Mouse Model to Test SARS-CoV-2 Viral Infection. RESEARCH SQUARE 2022:rs.3.rs-1431484. [PMID: 35475172 PMCID: PMC9040682 DOI: 10.21203/rs.3.rs-1431484/v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background: An animal model that can mimic the SARS-CoV-2 infection in humans is critical to understanding the rapidly evolving SARS-CoV-2 virus and for development of prophylactic and therapeutic strategies to combat emerging mutants. Studies show that the spike proteins of SARS-CoV and SARS-CoV-2 bind to human angiotensin-converting enzyme 2 (hACE2, a well-recognized, functional receptor for SARS-CoV and SARS-CoV-2) to mediate viral entry. Several hACE2 transgenic (hACE2Tg) mouse models are being widely used, which are clearly invaluable. However, the hACE2Tg mouse model cannot fully explain: 1) low expression of ACE2 observed in human lung and heart, but lung or heart failure occurs frequently in severe COVID-19 patients; 2) low expression of ACE2 on immune cells, but lymphocytopenia occurs frequently in COVID-19 patients; and 3) hACE2Tg mice do not mimic the natural course of SARS-CoV-2 infection in humans. Moreover, one of most outstanding features of coronavirus infection is the diversity of receptor usage, which includes the newly proposed human CD147 (hCD147) as a possible co-receptor for SARS-CoV-2 entry. It is still debatable whether CD147 can serve as a functional receptor for SARS-CoV-2 infection or entry. Results: Here we successfully generated a hCD147 knock-in mouse model (hCD147KI) in the NOD- scid IL2Rgamma null (NSG) background. In this hCD147KI-NSG mouse model, the hCD147 genetic sequence was placed downstream of the endogenous mouse promoter for mouse CD147 (mCD147), which creates an in vivo model that may better recapitulate physiological expression of hCD147 proteins at the molecular level compared to the existing and well-studied K18-hACE2-B6 (JAX) model. In addition, the hCD147KI-NSG mouse model allows further study of SARS-CoV-2 in the immunodeficiency condition which may assist our understanding of this virus in the context of high-risk populations in immunosuppressed states. Our data show 1) the human CD147 protein is expressed in various organs (including bronchiolar epithelial cells) in hCD147KI-NSG mice by immunohistochemical staining and flow cytometry; 2) hCD147KI-NSG mice are marginally sensitive to SARS-CoV-2 infection compared to WT-NSG littermates characterized by increased viral copies by qRT-PCR and moderate body weight decline compared to baseline; 3) a significant increase in leukocytes in the lungs of hCD147KI-NSG mice, compared to infected WT-NSG mice. Conclusions: hCD147KI-NSG mice are more sensitive to COVID-19 infection compared to WT-NSG mice. The hCD147KI-NSG mouse model can serve as an additional animal model for further interrogation whether CD147 serve as an independent functional receptor or accessory receptor for SARS-CoV-2 entry and immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Fei Chen
- Rutgers New Jersey Medical School
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Advanced drug delivery system against ischemic stroke. J Control Release 2022; 344:173-201. [PMID: 35248645 DOI: 10.1016/j.jconrel.2022.02.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 02/06/2023]
|
22
|
Zhang H, Zhao W. Resveratrol Alleviates Ischemic Brain Injury by Inhibiting the Activation of Pro-Inflammatory Microglia Via the CD147/MMP-9 Pathway. J Stroke Cerebrovasc Dis 2022; 31:106307. [PMID: 35093629 DOI: 10.1016/j.jstrokecerebrovasdis.2022.106307] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 12/24/2021] [Accepted: 01/04/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Ischemic stroke is one of the most common diseases with high mortality and disability. This study was intended to investigate the mechanism of resveratrol (RES) regulating microglia activation through the CD147/matrix metalloproteinase-9 (MMP-9) pathway on ischemic stroke. METHODS The middle cerebral artery occlusion (MCAO) mouse model and oxygen and glucose deprivation (OGD) cell model were established. The behavioral defects, neuronal damage, cerebral infarction volume, and histopathological changes were assessed in MCAO mice. The activation of pro-inflammatory microglia CD86+/Iba-1+ and anti-inflammatory microglia CD206+/Iba-1+ was detected. The expressions of pro-inflammatory microglia markers (CD11b, CD16) and cytokines (TNF-α, IL-1β, and IL-6) were measured. The activation of the CD147/MMP-9 pathway was detected and its effect on microglia activation was assessed. RESULTS After RES administration, the neuronal dysfunction, infarct volume, and morphological changes of neurons were improved in MCAO mice. Meanwhile, the motivation of pro-inflammatory microglia and the release of inflammatory factors were repressed. RES suppressed the stimulation of OGD/R microglia and the release of inflammatory factors. The expression of CD147 and MMP-9 in primary microglia was up-regulated. Inhibition of CD147 can reduce pro-inflammatory microglia activation by inhibiting MMP-9 expression. RES inhibited the CD147/MMP-9 axis in OGD/R microglia, and overexpression of CD147 partially reversed the inhibitory effect of RES on the activation and release of inflammatory factors in OGD/R microglia. CONCLUSION RES restrained the stimulation of pro-inflammatory microglia by down-regulating the CD147/MMP-9 axis, and thus protected against ischemic brain injury.
Collapse
Affiliation(s)
- Haifang Zhang
- Handan Emergency Rescue Command Center, Handan 056002 Hebei, China
| | - Wenjing Zhao
- Department of Neurology, The Affiliated Hospital of Hebei University of Engineering, Handan 056002 Hebei, China.
| |
Collapse
|
23
|
Finger CE, Moreno-Gonzalez I, Gutierrez A, Moruno-Manchon JF, McCullough LD. Age-related immune alterations and cerebrovascular inflammation. Mol Psychiatry 2022; 27:803-818. [PMID: 34711943 PMCID: PMC9046462 DOI: 10.1038/s41380-021-01361-1] [Citation(s) in RCA: 86] [Impact Index Per Article: 28.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 09/20/2021] [Accepted: 10/12/2021] [Indexed: 12/11/2022]
Abstract
Aging is associated with chronic systemic inflammation, which contributes to the development of many age-related diseases, including vascular disease. The world's population is aging, leading to an increasing prevalence of both stroke and vascular dementia. The inflammatory response to ischemic stroke is critical to both stroke pathophysiology and recovery. Age is a predictor of poor outcomes after stroke. The immune response to stroke is altered in aged individuals, which contributes to the disparate outcomes between young and aged patients. In this review, we describe the current knowledge of the effects of aging on the immune system and the cerebral vasculature and how these changes alter the immune response to stroke and vascular dementia in animal and human studies. Potential implications of these age-related immune alterations on chronic inflammation in vascular disease outcome are highlighted.
Collapse
Affiliation(s)
- Carson E. Finger
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA
| | - Ines Moreno-Gonzalez
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA ,grid.10215.370000 0001 2298 7828Department of Cell Biology, Genetics and Physiology, Instituto de Investigacion Biomedica de Malaga-IBIMA, Faculty of Sciences, Malaga University, Malaga, Spain ,grid.418264.d0000 0004 1762 4012Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Malaga, Spain
| | - Antonia Gutierrez
- grid.10215.370000 0001 2298 7828Department of Cell Biology, Genetics and Physiology, Instituto de Investigacion Biomedica de Malaga-IBIMA, Faculty of Sciences, Malaga University, Malaga, Spain ,grid.418264.d0000 0004 1762 4012Biomedical Research Networking Center on Neurodegenerative Diseases (CIBERNED), Malaga, Spain
| | - Jose Felix Moruno-Manchon
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, UTHealth Science Center at Houston, Houston, TX USA
| |
Collapse
|
24
|
Neuroprotection of minocycline by inhibition of extracellular matrix metalloproteinase inducer expression following intracerebral hemorrhage in mice. Neurosci Lett 2021; 764:136297. [PMID: 34666120 DOI: 10.1016/j.neulet.2021.136297] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 09/30/2021] [Accepted: 10/13/2021] [Indexed: 12/25/2022]
Abstract
Intracerebral hemorrhage (ICH) is a severe neurological dysfunction and a medical emergency with a high mortality rate. Minocycline ameliorates deficits in rodent models of acute and chronic neurological diseases. However, the role of minocycline in ICH remains unclear. The extracellular matrix metalloproteinase inducer (EMMPRIN) is a key inflammatory mediator in some neurological diseases, triggering matrix metalloproteinases (MMPs) production. In this study, we aimed to use minocycline to inhibit EMMPRIN and thus the activity of MMPs. Male adult C57BL/6 mice were injected with collagenase type VII or saline into the right basal ganglia and euthanized at different time points. The minocycline was intraperitoneally injected once every 12 h for three days to block the expression of EMMPRIN from two hours after ICH. We found that breakdown of the BBB was most severe 3 days after ICH. The minocycline treatment significantly decreased EMMPRIN and MMP-9 expression, reduced zonula occludens-1 and occludin, and alleviated BBB disruption. Moreover, minocycline treatment displayed a lower brain water content, lesser neurological dysfunction, and smaller injury volume on day 3 than those of the vehicle-treated group. Minocycline also inhibited the activation of microglia/macrophages, infiltration of neutrophils, and production of inflammatory mediators, including tumor necrosis factor alpha and interleukin-1beta. The current study shows that minocycline exhibits protective roles in ICH by decreasing EMMPRIN and MMP-9 expression, alleviating BBB disruption, inhibiting neuroinflammation, areducing neuronal degeneration and death.
Collapse
|
25
|
Wang H, Lv J, Zhao Y, Wei H, Zhang T, Yang H, Chen Z, Jiang J. Endothelial genetic deletion of CD147 induces changes in the dual function of the blood-brain barrier and is implicated in Alzheimer's disease. CNS Neurosci Ther 2021; 27:1048-1063. [PMID: 33987940 PMCID: PMC8339530 DOI: 10.1111/cns.13659] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 04/20/2021] [Accepted: 04/24/2021] [Indexed: 12/19/2022] Open
Abstract
AIMS The blood-brain barrier (BBB) is a specialized and indispensable structure in brain blood vessels that is damaged during Alzheimer's disease (AD). CD147 is expressed on the BBB and deeply engaged in the AD pathological process. In this study, we aimed to provide a better understanding of the roles of CD147 in BBB function in health and neurodegenerative disease. METHODS AND RESULTS We measured CD147 expression in mouse brains and demonstrated that CD147 is exclusively expressed in brain endothelial cells (BECs), and its expression decreases with age. After constructing endothelial-specific CD147 knockout mice, we performed RNA-sequencing on BECs isolated from mice of different ages as well as a range of database analyses. We found that endothelial CD147 is essential for the dual functions of the BBB, including barrier maintenance and transporter regulation. This study also shows that CD147 plays a pivotal role in neurodegenerative diseases, particularly in AD. CONCLUSIONS Our findings suggested that targeting CD147 in BECs may represent a novel therapeutic strategy, which promoted the design of future experimental investigations and the mechanistic understanding of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hao Wang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Jian‐Jun Lv
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Yu Zhao
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Hao‐Lin Wei
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Tian‐Jiao Zhang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Hai‐Jiao Yang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Zhi‐Nan Chen
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| | - Jian‐Li Jiang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi’anChina
| |
Collapse
|
26
|
Badeti S, Tseng HC, Romanienko P, Yehia G, Liu D. Development of a Novel Human CD147 Transgenic NSG Mouse Model to test SARS-CoV-2 Infection and Immune Responses. RESEARCH SQUARE 2021. [PMID: 33851148 PMCID: PMC8043462 DOI: 10.21203/rs.3.rs-396257/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
An animal model that can mimic the SARS-CoV-2 infection in humans is critical to understanding the newly emerged, rapidly spreading SARS-CoV-2 and development of therapeutic strategies. Studies show that the spike (S) proteins of SARS-CoV (SARS-CoV-S-1-S) and SARS-CoV-2 (SARS-CoV-2-S) bind to human angiotensin-converting enzyme 2 (hACE2, a well-recognized, functional receptor for SARS-CoV and SARS-CoV-2) to mediate viral entry. Several hACE2 transgenic (hACE2Tg) mouse models are being widely used, which is clearly invaluable. However, the hACE2Tg mouse model cannot fully explain: 1) low expression of ACE2 observed in human lung and heart, but lung or heart failure occurs frequently in severe COVID-19 patients); 2) low expression of ACE2 on immune cells, but lymphocytopenia occurs frequently in COVID-19 patients; and 3) hACE2Tg mice do not develop strong clinical disease following SARS-CoV-2 infection in contrast to SARS-CoV-1. Moreover, one of most outstanding features of coronaviruses is the diversity of receptor usage, which includes the newly proposed human CD147 (hCD147) as a receptor for SARS-CoV-2-S. It is still debatable whether CD147 can serve as a functional receptor for SARS-CoV-2 infection or entry. Here we successfully generated a hCD147Tg mouse model in the NOD-scid IL2Rgammanull (NSG) background. In this hCD147Tg-NSG mouse model, the hCD147 genetic sequence was placed following the endogenous mouse promoter for mouse CD147 (mCD147), which creates an in vivo model that may better recapitulate physiological expression of CD147 proteins at the molecular level compared to the existing and well-studied K18-hACE2-B6 model. In addition, the hCD147Tg-NSG mouse model allows further study of SARS-CoV-2 in the immunodeficiency condition which may assist our understanding of this virus in the context of high-risk populations with immunosuppressed conditions. The hCD147Tg-NSG mouse mode can serve as an additional animal model for interrogate whether CD147 serve as an independent functional receptor or accessory receptor for SARS-CoV-2 entry and immune responses.
Collapse
|
27
|
Zhi C, Zeng S, Chen Y, Liao D, Lai M, Wang Z, Wang Y, Xiao S. Clemastine promotes recovery of neural function and suppresses neuronal apoptosis by restoring balance of pro-inflammatory mediators in an experimental model of intracerebral hemorrhage. Int J Med Sci 2021; 18:639-645. [PMID: 33437198 PMCID: PMC7797547 DOI: 10.7150/ijms.51150] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/17/2020] [Indexed: 11/19/2022] Open
Abstract
Intracerebral hemorrhage (ICH) represents a common acute cerebrovascular event that imparts high rates of disability. The microglia-mediated inflammatory response is a critical factor in determining cerebral damage post-ICH. Clemastine (CLM) is a histamine receptor H1 (HRH1) antagonist that has been shown to modulate the inflammatory response. However, the effects of CLM on ICH and the underlying mechanism remain to be determined. This investigation reveals that CLM resulted in reduction of cerebral hematoma volume, decreased cerebral edema and lower rates of neuronal apoptosis as well as improved behavioral scores in an acute ICH murine model. CLM treatment was noted to decrease pro-inflammatory effectors and increased anti-inflammatory effectors post-ICH. In addition, CLM reduced the deleterious effects of activated microglia on neurons in a transwell co-culture system. Our findings show that CLM likely mediates its therapeutic effect through inhibition of microglia-induced inflammatory response and apoptosis, thereby enhancing restoration of neuronal function.
Collapse
Affiliation(s)
- Cheng Zhi
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Shulian Zeng
- Department of Neurology, Heyuan People's Hospital, Heyuan, 517000 China
| | - Yuan Chen
- Department of Respiratory and Critical Care Medicine, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Degui Liao
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Miaoling Lai
- Department of Pathology, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Zhaotao Wang
- Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260 China
| | - Yezhong Wang
- Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260 China
| | - Shiyin Xiao
- Institute of Neuroscience, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260 China
| |
Collapse
|
28
|
Cohen-Salmon M, Slaoui L, Mazaré N, Gilbert A, Oudart M, Alvear-Perez R, Elorza-Vidal X, Chever O, Boulay AC. Astrocytes in the regulation of cerebrovascular functions. Glia 2020; 69:817-841. [PMID: 33058289 DOI: 10.1002/glia.23924] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 09/18/2020] [Accepted: 09/21/2020] [Indexed: 12/18/2022]
Abstract
Astrocytes are the most numerous type of neuroglia in the brain and have a predominant influence on the cerebrovascular system; they control perivascular homeostasis, the integrity of the blood-brain barrier, the dialogue with the peripheral immune system, the transfer of metabolites from the blood, and blood vessel contractility in response to neuronal activity. These regulatory processes occur in a specialized interface composed of perivascular astrocyte extensions that almost completely cover the cerebral blood vessels. Scientists have only recently started to study how this interface is formed and how it influences cerebrovascular functions. Here, we review the literature on the astrocytes' role in the regulation of the cerebrovascular system. We cover the anatomy and development of the gliovascular interface, the known gliovascular functions, and molecular factors, the latter's implication in certain pathophysiological situations, and recent cutting-edge experimental tools developed to examine the astrocytes' role at the vascular interface. Finally, we highlight some open questions in this field of research.
Collapse
Affiliation(s)
- Martine Cohen-Salmon
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Leila Slaoui
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Noémie Mazaré
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Alice Gilbert
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Marc Oudart
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Rodrigo Alvear-Perez
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Xabier Elorza-Vidal
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| | - Oana Chever
- Normandie University, UNIROUEN, INSERM, DC2N, IRIB, Rouen, France
| | - Anne-Cécile Boulay
- Physiology and Physiopathology of the Gliovascular Unit Research Group, Center for Interdisciplinary Research in Biology (CIRB), College de France, CNRS Unité Mixte de Recherche 724, INSERM Unité 1050, Labex Memolife, PSL Research University, Paris, France
| |
Collapse
|