1
|
Ball BK, Park JH, Bergendorf AM, Proctor EA, Brubaker DK. Translational disease modeling of peripheral blood identifies type 2 diabetes biomarkers predictive of Alzheimer's disease. NPJ Syst Biol Appl 2025; 11:58. [PMID: 40442087 PMCID: PMC12122922 DOI: 10.1038/s41540-025-00539-5] [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: 01/09/2025] [Accepted: 05/16/2025] [Indexed: 06/02/2025] Open
Abstract
Type 2 diabetes (T2D) is a significant risk factor for Alzheimer's disease (AD). Despite multiple studies reporting this connection, the mechanism by which T2D exacerbates AD is poorly understood. It is challenging to design studies that address co-occurring and comorbid diseases, limiting the number of existing evidence bases. To address this challenge, we expanded the applications of a computational framework called Translatable Components Regression (TransComp-R), initially designed for cross-species translation modeling, to perform cross-disease modeling to identify biological programs of T2D that may exacerbate AD pathology. Using TransComp-R, we combined peripheral blood-derived T2D and AD human transcriptomic data to identify T2D principal components predictive of AD status. Our model revealed genes enriched for biological pathways associated with inflammation, metabolism, and signaling pathways from T2D principal components predictive of AD. The same T2D PC predictive of AD outcomes unveiled sex-based differences across the AD datasets. We performed a gene expression correlational analysis to identify therapeutic hypotheses tailored to the T2D-AD axis. We identified six T2D and two dementia medications that induced gene expression profiles associated with a non-T2D or non-AD state. We next assessed our blood-based T2DxAD biomarker signature in post-mortem human AD and control brain gene expression data from the hippocampus, entorhinal cortex, superior frontal gyrus, and postcentral gyrus. Using partial least squares discriminant analysis, we identified a subset of genes from our cross-disease blood-based biomarker panel that significantly separated AD and control brain samples. Finally, we validated our findings using single cell RNA-sequencing blood data of AD and healthy individuals and found erythroid cells contained the most gene expression signatures to the T2D PC. Our methodological advance in cross-disease modeling identified biological programs in T2D that may predict the future onset of AD in this population. This, paired with our therapeutic gene expression correlational analysis, also revealed alogliptin, a T2D medication that may help prevent the onset of AD in T2D patients.
Collapse
Affiliation(s)
- Brendan K Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Alexander M Bergendorf
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Elizabeth A Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Penn State Neuroscience Institute, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA.
| |
Collapse
|
2
|
Chen C, Xu X, Lu J, Xiang Y, Shi L, Liu D. Hyperglycemia-induced blood-brain barrier dysfunction: Mechanisms and therapeutic interventions. Microvasc Res 2025; 160:104820. [PMID: 40393562 DOI: 10.1016/j.mvr.2025.104820] [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: 03/04/2025] [Revised: 04/09/2025] [Accepted: 05/14/2025] [Indexed: 05/22/2025]
Abstract
The blood-brain barrier (BBB) serves as a highly selective interface that regulates the transport of molecules between the blood and the brain. Its integrity is essential for maintaining neuronal homeostasis and preventing neuroinflammation. Hyperglycemia, a hallmark of diabetes, is linked to cognitive deficits and central nervous system (CNS) pathologies, including vascular dementia, stroke, and Alzheimer's disease, with BBB damage as a potential contributing factor. As the global prevalence of diabetes rises, understanding the connection between hyperglycemia and BBB dysfunction may facilitate the development of novel treatments that protect or restore BBB integrity, thereby alleviating the neurological complications of diabetes. Furthermore, it may aid in the development of targeted therapies for diabetes-related neurological complications. This literature review examines the emerging insights into the relationship between hyperglycemia and BBB dysfunction. It focuses on the mechanisms underlying BBB dysfunction, the clinical manifestations of this dysfunction in diabetes and cerebrovascular diseases, and potential therapeutic interventions.
Collapse
Affiliation(s)
- Changsheng Chen
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, Jiangsu Province, China.
| | - Xi Xu
- Medical College of Nantong University, Nantong, Jiangsu Province, China
| | - Jiahao Lu
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, Jiangsu Province, China
| | - Yuqing Xiang
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, Jiangsu Province, China
| | - Linsheng Shi
- Department of Cardiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province, China
| | - Dong Liu
- School of Life Sciences, Nantong Laboratory of Development and Diseases, Nantong University, Nantong, Jiangsu Province, China; Medical College of Nantong University, Nantong, Jiangsu Province, China; Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong, Jiangsu Province, China.
| |
Collapse
|
3
|
Menet R, Nasrallah L, Bernard M, Allain A, ElAli A. VEGF-E Attenuates Injury After Ischemic Stroke by Promoting Reparative Revascularization. Eur J Neurosci 2025; 61:e70114. [PMID: 40277075 PMCID: PMC12023035 DOI: 10.1111/ejn.70114] [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: 11/21/2024] [Revised: 03/21/2025] [Accepted: 04/07/2025] [Indexed: 04/26/2025]
Abstract
The angiogenic response after stroke correlates with mild injury and an improved recovery. Stimulation of post-stroke angiogenesis using vascular endothelial growth factor (VEGF)-A is associated with an increased risk of vascular destabilization, leading to life-threatening complications. The non-mammalian VEGF-A homolog, VEGF-E, stimulates stable cutaneous vascularization and promotes wound healing. Herein, we posit that VEGF-E represents a potential mediator of reparative revascularization after ischemic stroke. C57BL6/J wildtype mice were subjected to experimental stroke, and VEGF-E or VEGF-A were intranasally delivered during the subacute phase. Our results indicate that VEGF-E improves neurological recovery and increases vascular density without compromising permeability, more efficiently than VEGF-A. We show that VEGF-E-mediated revascularization correlates with normal restoration of brain perfusion, whereas VEGF-A induces cerebral hyperperfusion, indicative of vascular dysfunction. Furthermore, VEGF-E reduces microvascular stalls, increases the density of angiogenic vasculature, and improves the interaction of brain endothelial cell with pericytes, which is critical for vascular stabilization. Using cell-based assays, we demonstrate that stimulation of brain endothelial cells with VEGF-E, but not with VEGF-A, increases the expression of platelet-derived growth factor (PDGF)-D, a potent ligand of PDGFRβ that plays critical roles in regulating the survival and functions of perivascular cells, including pericytes. These effects are associated with activation of extracellular signal-regulated kinase (ERK)1/2 and P38 mitogen-activated protein kinase (MAPK). Finally, we confirm that the secretome of VEGF-E-stimulated brain endothelial cells ameliorates pericyte migration required for vascular recruitment. Our study indicates that VEGF-E promotes a stable and functional revascularization after ischemic stroke, outlining its promises for therapeutic purposes.
Collapse
Affiliation(s)
- Romain Menet
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Leila Nasrallah
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Maxime Bernard
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Anne‐Sophie Allain
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| | - Ayman ElAli
- Department of Psychiatry and Neuroscience, Faculty of MedicineUniversité LavalQuebec CityQuebecCanada
- Neuroscience AxisResearch Center of CHU de Québec ‐ Université LavalQuebec CityQuebecCanada
| |
Collapse
|
4
|
Agrawal N, Afzal M, Khan NH, Ganesan S, Kumari M, Sunitha S, Dash A, Goyal K, Kushwaha B, Rekha A, Rana M, Ali H. The role of VEGF in vascular dementia: impact of aging and cellular senescence. Biogerontology 2025; 26:77. [PMID: 40119956 DOI: 10.1007/s10522-025-10219-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025]
Abstract
Vascular Endothelial Growth Factor (VEGF) is a critical element in vascular dementia (VD) pathogenesis and therapeutic development while remaining strongly influenced by aging processes and cellular aging mechanisms. VEGF's multiple effects comprise neuroprotective functions, its role in vascular development, and its ability to regulate brain blood flow systems, all leading to cognitive preservation. The prefrontal cortex exhibits elevated VEGF gene levels, which directly matches the advancement of cognitive deficits in patients with Alzheimer's disease and VD. These patients exhibit higher VEGF levels in their CSF fluid, demonstrating that disease pathology includes multiple inseparable factors. Aging dramatically worsens VEGF regulation because endothelial dysfunction combines with chronic inflammation and oxidative stress to generate adverse vascular symptoms that include atherosclerosis and stroke. Cellular senescence convolutes these processes by causing damaging inflammatory reactions alongside impaired vascular healing abilities. The secretion of pro-inflammatory cytokines from senescent cells (SCs) disrupts VEGF signaling and produces harmful consequences for both vascular health and cognitive well-being. The neuroprotective properties of VEGF-A165a and VEGF-A165b variants demonstrate their ability to lessen β-amyloid and tau protein toxicity. The protective mechanisms of VEGF depend heavily on VEGF expression levels and receptor functionality, both of which decrease with aging. The combination of approaches that modulate VEGF signaling and SC accumulation shows potential for designing treatments against VD. People can sustain BBB functionality over a longer period through Mediterranean diet consumption together with aerobic exercise along with developing therapies, including senolytics and senomorphics, which delay neurodegenerative progression. Future investigative efforts must improve VEGF delivery methods while studying cellular senescence mechanisms and developing advanced methods to detect SC cells. A three-dimensional healthcare approach combining VEGF-targeted treatments with anti-ageing interventions and detailed diagnostic techniques shows the potential for effective VD management to achieve better patient results.
Collapse
Affiliation(s)
- Neetu Agrawal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah, 21442, Saudi Arabia
| | - Nawaid Hussain Khan
- Faculty of Medicine, Ala-Too International University, Bishkek, Kyrgyz Republic.
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mukesh Kumari
- Department of Applied Sciences-Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, India
| | - S Sunitha
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Aniruddh Dash
- Department of Orthopaedics, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun, 248002, India
| | - Brajgopal Kushwaha
- IES Institute of Pharmacy, IES University, Bhopal, Madhya Pradesh, 462044, India
| | - A Rekha
- Hospital and Research Centre, Dr. D. Y. Patil Medical College, Pimpri, Pune, India
| | - Mohit Rana
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Institute of Medical and Technical Sciences, Saveetha Medical College,, Saveetha University, Chennai, India
| |
Collapse
|
5
|
Nian Y, Hu FY, Wang YM, Yue WJ, Xie Y, Li B, Zhou LH. Continuity of care and 5E nursing intervention improve neurological function, quality of life, and prognosis for acute ischemic stroke patients. Am J Transl Res 2025; 17:1694-1706. [PMID: 40225997 PMCID: PMC11982865 DOI: 10.62347/ubva3419] [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: 09/28/2024] [Accepted: 02/09/2025] [Indexed: 04/15/2025]
Abstract
OBJECTIVE To evaluate the effect of continuous nursing combined with comprehensive 5E rehabilitation-based nursing intervention on neurological function, quality of life, and prognosis in patients with acute ischemic stroke (AIS). METHODS This retrospective study included 117 AIS patients admitted to TaiHe County People's Hospital between January 2022 and March 2024. Patients receiving routine nursing care from January 2022 to March 2023 were assigned to the conventional care group (56 cases), while those receiving continuous nursing combined with comprehensive 5E rehabilitation-based nursing intervention from March 2023 to March 2024 were assigned to the integrated care group (61 cases). Pre- and post-intervention comparisons were made between the groups regarding neurological function, cognitive ability, physical capability, coagulation status, activities of daily living, quality of life, nursing satisfaction, and prognosis. Additionally, risk factors for poor prognosis in AIS patients were analyzed based on their outcomes. RESULTS Prior to intervention, there were no significant differences in neurological function, cognitive function, physical function, coagulation status, daily living ability, or quality of life scores between the two groups (all P>0.05). After intervention, the integrated care group showed significantly better outcomes in neurological function, cognitive function, physical function, coagulation status, daily living ability, quality of life, nursing satisfaction, and prognosis compared to the conventional care group (all P<0.05). Multivariate logistic regression analysis identified older age, high National Institutes of Health Stroke Scale score, elevated serum C-reactive protein and homocysteine levels, and reduced vascular endothelial growth factor levels as risk factors for poor prognosis in AIS patients (all P<0.05). CONCLUSION The integration of continuity of care with comprehensive nursing intervention based on the 5E rehabilitation framework effectively addresses the rehabilitation needs of AIS patients, significantly improving neurological function, quality of life, and prognosis.
Collapse
Affiliation(s)
- Yu Nian
- Department of Neurology, Taihe County People’s HospitalFuyang 236600, Anhui, China
| | - Feng-Ying Hu
- Department of Neurology, Taihe County People’s HospitalFuyang 236600, Anhui, China
| | - Yan-Mei Wang
- Department of Neurology, Taihe County People’s HospitalFuyang 236600, Anhui, China
| | - Wen-Jing Yue
- Department of Neurology, Taihe County People’s HospitalFuyang 236600, Anhui, China
| | - Yun Xie
- Department of Neurology, Taihe County People’s HospitalFuyang 236600, Anhui, China
| | - Bo Li
- Department of Neurology, Taihe County People’s HospitalFuyang 236600, Anhui, China
| | - Li-Heng Zhou
- Department of Nursing, Taihe County People’s HospitalFuyang 236600, Anhui, China
| |
Collapse
|
6
|
Guo J, Wang G, Liu T, Zhang J, Li Q, Zhu Y, Luo H. Acupuncture Improves Chronic Cerebral Ischemia by Inhibiting the CKLF1/HIF-1α/VEGF/Notch1 Signaling Pathway. CNS Neurosci Ther 2025; 31:e70246. [PMID: 40019048 PMCID: PMC11868988 DOI: 10.1111/cns.70246] [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: 09/26/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE Acupuncture significantly improves cognitive dysfunction in rats with chronic cerebral ischemia. However, the underlying signaling pathways remain unclear. This study investigates the role of the CKLF1/HIF-1α/VEGF/Notch1 signaling pathway in the acupuncture-mediated improvement of cognitive function in rats with chronic cerebral ischemia. METHODS Male SD rats were randomly divided into the normal control group, sham-operated group, 2-VO model group, 2-VO + acupuncture group, and 2-VO + Ginaton group (Ginkgo biloba extract 14.4 mg/kg/day), with 10 rats in each group. The 2-VO + acupuncture group received acupuncture at the Shuigou, Baihui, bilateral Fengchi, and bilateral Zusanli points for 14 consecutive sessions over 2 weeks. The rats' memory function was assessed using the Morris water maze and novel object recognition tests. Cerebral blood volume changes were measured using laser speckle imaging. Ultrastructural changes in microvessels were observed via transmission electron microscopy. Neuronal and myelin alterations were evaluated using HE staining, Nissl staining, and LFB myelin staining. The expression levels of CKLF1, CCR5, HIF-1α, VEGF, and Notch1 proteins were measured using Western blot, and multiple immunofluorescence staining was performed to assess the colocalization of CKLF1 and neurons. RESULTS Compared with the 2-VO model group, acupuncture treatment reduced the latency period and increased the number of platform crossings in the Morris water maze test, and the 2-VO model group had a higher recognition index in the novel object recognition test. We found that acupuncture improved the condition of endothelial cells, repaired the morphology of the vascular lumen, and alleviated astrocyte edema. We also showed that acupuncture could ameliorate pathological damage in rats with chronic cerebral ischemia. Moreover, acupuncture reduced the expression of CKLF1, CCR5, and HIF-1α proteins in the hippocampus, decreased the fluorescence intensity of CKLF1 expression, and increased the fluorescence intensity of neurons in the hippocampal CA1 region. CONCLUSION Acupuncture may exert neuroprotective effects and improve cognitive dysfunction caused by chronic cerebral ischemia by regulating the CKLF1/HIF-1α/VEGF/Notch1 pathway to inhibit inflammatory factors and increase cerebral blood flow.
Collapse
Affiliation(s)
- Jilong Guo
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- Jinzhong City Hospital of Traditional Chinese MedicineJinzhongChina
| | - Guangqi Wang
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- National International Joint Research Center for Molecular Traditional Chinese MedicineJinzhongChina
| | - Tian Liu
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- National International Joint Research Center for Molecular Traditional Chinese MedicineJinzhongChina
| | - Jianjun Zhang
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- National International Joint Research Center for Molecular Traditional Chinese MedicineJinzhongChina
| | - Qinqing Li
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- National International Joint Research Center for Molecular Traditional Chinese MedicineJinzhongChina
| | - Yousong Zhu
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- National International Joint Research Center for Molecular Traditional Chinese MedicineJinzhongChina
| | - Huijuan Luo
- Shanxi Key Laboratory of Chinese Medicine EncephalopathyJinzhongChina
- Basic Medical College of Shanxi University of Chinese MedicineJinzhongChina
- National International Joint Research Center for Molecular Traditional Chinese MedicineJinzhongChina
| |
Collapse
|
7
|
Kanayama T, Hatakeyama M, Akiyama N, Otsu Y, Onodera O, Shimohata T, Kanazawa M. Oxygen-glucose-deprived peripheral blood mononuclear cells act on hypoxic lesions after ischemia-reperfusion injury. Exp Neurol 2025; 385:115121. [PMID: 39710242 DOI: 10.1016/j.expneurol.2024.115121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 12/14/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Despite advances in reperfusion therapies, ischemic stroke remains a major cause of long-term disability due to residual hypoxic lesions persisting after macrovascular reperfusion. These residual hypoxic lesions, caused by microvascular dysfunction, represent an important therapeutic target. We previously demonstrated that oxygen-glucose-deprived peripheral blood mononuclear cells (OGD-PBMCs) migrate to ischemic brain regions and promote functional recovery after stroke. This recovery occurs through mechanisms involving hypoxia-inducible factor-1α, exosomal miR-155-5p, and vascular endothelial growth factor (VEGF). However, it remains unclear whether OGD-PBMCs target hypoxic regions. METHODS We evaluated cerebral blood flow using a laser speckle flow imaging system. Next, we utilized pimonidazole to investigate the presence of hypoxic lesions after ischemia-reperfusion injury in a rat suture occlusion model in immunohistochemical analyses. We also compared levels of a cell surface receptor in human PBMCs by flow cytometric analysis under normoxic and OGD conditions. RESULTS We found persistent pimonidazole-positive hypoxic lesions at 10- and 28-days post-reperfusion despite restored gross cerebral perfusion. Treatment with the C-X-C motif chemokine receptor 4 (CXCR4) inhibitor AMD3100 before and after OGD-PBMCs administration reduced the number of OGD-PBMCs in the brain parenchyma compared to the control group (P = 0.018). Administered OGD-PBMCs localized within these hypoxic regions via the stromal cell-derived factor-1/CXCR4 chemotactic axis. OGD-PBMCs enhanced VEGF expression, specifically within hypoxic lesions, compared to the phosphate-buffered saline group (P < 0.01). Furthermore, OGD-PBMCs reduced the number of pimonidazole-positive hypoxic cells in the ischemic core on 28 days. These findings demonstrate that OGD-PBMCs selectively migrate to and modulate the microenvironment of hypoxic lesions following cerebral ischemia-reperfusion injury. CONCLUSION Targeting these residual hypoxic regions may underline the therapeutic effects of OGD-PBMC treatment and represent a promising strategy for improving stroke recovery despite successful recanalization.
Collapse
Affiliation(s)
- Takeshi Kanayama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata 951-8585, Japan
| | - Masahiro Hatakeyama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata 951-8585, Japan
| | - Natsuki Akiyama
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata 951-8585, Japan
| | - Yutaka Otsu
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata 951-8585, Japan
| | - Osamu Onodera
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata 951-8585, Japan
| | - Takayoshi Shimohata
- Department of Neurology, Gifu University Graduate School of Medicine, 1-1 Yanagido, Gifu 501-1194, Japan
| | - Masato Kanazawa
- Department of Neurology, Brain Research Institute, Niigata University, 1-757 Asahimachi-dori, Chuoku, Niigata 951-8585, Japan.
| |
Collapse
|
8
|
Grygorczuk S, Czupryna P, Martonik D, Adamczuk J, Parfieniuk-Kowerda A, Grzeszczuk A, Pawlak-Zalewska W, Dunaj-Małyszko J, Mielczak K, Parczewski M, Moniuszko-Malinowska A. The Factors Associated with the Blood-Brain Barrier Dysfunction in Tick-Borne Encephalitis. Int J Mol Sci 2025; 26:1503. [PMID: 40003967 PMCID: PMC11855613 DOI: 10.3390/ijms26041503] [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: 12/17/2024] [Revised: 01/31/2025] [Accepted: 02/09/2025] [Indexed: 02/27/2025] Open
Abstract
The pathogenesis of the central nervous system (CNS) pathology in tick-borne encephalitis (TBE) remains unclear. We attempted to identify mediators of the blood-brain barrier (BBB) disruption in human TBE in paired serum and cerebrospinal fluid (CSF) samples from 100 TBE patients. CSF albumin quotient (Qalb) was calculated as a measure of BBB impairment. Concentrations of cytokines, cytokine antagonists, adhesion molecules, selectins and matrix metalloproteinases (MMP) were measured with a multiplex bead assay. Single nucleotide polymorphisms (SNP) in genes MIF, TNF, TNFRSF1A, TNFRSF1B, IL-10, TLR3 and TLR4 were studied in patient blood DNA extracts and analyzed for associations with Qalb and/or cytokine concentrations. The multivariate regression models of Qalb were built with the soluble mediators as independent variables. The best models obtained included L-selectin, P-selectin, sVCAM, MMP7, MMP8 (or MMP9) and IL-28A as positive and IL-12p70, IL-15, IL-6Rα/IL-6 ratio and TNF-RII/TNFα ratio as negative correlates of Qalb. The genotype did not associate with Qalb, but polymorphism rs4149570 (in TNFRSF1A) associated with TNFα and rs1800629 (TNF) with MIF concentration. We confirm the association of the TNFα-dependent response, L-selectin and MMP8/MMP9 with BBB disruption and identify its novel correlates (IL-12, IL-15, IL-28A, MMP7). We detect no genotype associations with BBB function in TBE.
Collapse
Affiliation(s)
- Sambor Grygorczuk
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| | - Piotr Czupryna
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| | - Diana Martonik
- Department of Infectious Diseases and Hepatology, Medical University in Białystok, 15-089 Białystok, Poland; (D.M.); (A.P.-K.)
| | - Justyna Adamczuk
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| | - Anna Parfieniuk-Kowerda
- Department of Infectious Diseases and Hepatology, Medical University in Białystok, 15-089 Białystok, Poland; (D.M.); (A.P.-K.)
| | - Anna Grzeszczuk
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| | - Wioletta Pawlak-Zalewska
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| | - Justyna Dunaj-Małyszko
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| | - Kaja Mielczak
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University, 70-204 Szczecin, Poland; (K.M.); (M.P.)
| | - Miłosz Parczewski
- Department of Infectious, Tropical Diseases and Acquired Immunodeficiency, Pomeranian Medical University, 70-204 Szczecin, Poland; (K.M.); (M.P.)
| | - Anna Moniuszko-Malinowska
- Department of Infectious Diseases and Neuroinfections, Medical University in Białystok, 15-089 Białystok, Poland; (P.C.); (J.A.); (A.G.); (W.P.-Z.); (J.D.-M.); (A.M.-M.)
| |
Collapse
|
9
|
Sprincl V, Romanyuk N. miRNA in blood-brain barrier repair: role of extracellular vesicles in stroke recovery. Front Cell Neurosci 2025; 19:1503193. [PMID: 39990970 PMCID: PMC11842324 DOI: 10.3389/fncel.2025.1503193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/24/2025] [Indexed: 02/25/2025] Open
Abstract
Ischemic stroke is a leading cause of mortality and long-term disability globally. One of its aspects is the breakdown of the blood-brain barrier (BBB). The disruption of BBB's integrity during stroke exacerbates neurological damage and hampers therapeutic intervention. Recent advances in regenerative medicine suggest that mesenchymal stem cells (MSCs) derived extracellular vesicles (EVs) show promise for restoring BBB integrity. This review explores the potential of MSC-derived EVs in mediating neuroprotective and reparative effects on the BBB after ischemic stroke. We highlight the molecular cargo of MSC-derived EVs, including miRNAs, and their role in enhancing angiogenesis, promoting the BBB and neural repair, and mitigating apoptosis. Furthermore, we discuss the challenges associated with the clinical translation of MSC-derived EV therapies and the possibilities of further enhancing EVs' innate protective qualities. Our findings underscore the need for further research to optimize the therapeutic potential of EVs and establish their efficacy and safety in clinical settings.
Collapse
Affiliation(s)
- Vojtech Sprincl
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
- Department of Neuroscience, 2nd Medical Faculty, Charles University, Prague, Czechia
| | - Nataliya Romanyuk
- Department of Neuroregeneration, Institute of Experimental Medicine of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
10
|
Gu CL, Zhang L, Zhu Y, Bao TY, Zhu YT, Chen YT, Pang HQ. Exploring the cellular and molecular basis of nerve growth factor in cerebral ischemia recovery. Neuroscience 2025; 566:190-197. [PMID: 39742942 DOI: 10.1016/j.neuroscience.2024.12.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/04/2025]
Abstract
Vascular obstruction often causes inadequate oxygen and nutrient supply to the brain. This deficiency results in cerebral ischemic injury, which significantly impairs neurological function. This review aimed to explore the neuroprotective and regenerative effects of nerve growth factor (NGF) in cerebral ischemic injury. NGF, a crucial neurotrophic factor, could inhibit neuronal apoptosis, reduce inflammatory responses, and promote axon regeneration and angiogenesis through its interaction with TrkA, a high-affinity receptor. These functions were closely related to the activation of Phosphatidylinositol 3-kinase/Protein kinase B (PI3K/AKT) and Mitogen-Activated Protein Kinase (MAPK) pathways. Moreover, the mechanisms of NGF in the acute and recovery phases, along with the strategies to enhance its therapeutic effects using delivery systems (such as intranasal administration, nanovesicles, and gene therapy) were also summarized. Although NGF shows great potential for clinical application, its delivery efficiency and long-term safety still need more research and improvements. Future research should focus on exploring the specific action mechanism of NGF, optimizing the delivery strategy, and evaluating its long-term efficacy and safety to facilitate its clinical transformation in cerebral ischemic stroke.
Collapse
Affiliation(s)
- Chen-Lin Gu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Lu Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan 030001, China
| | - Yan Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Ting-Yu Bao
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Ting Zhu
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Yu-Tong Chen
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China
| | - Han-Qing Pang
- Institute of Translational Medicine, School of Medicine, Yangzhou University, Yangzhou 225009, China; Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou 225009, China.
| |
Collapse
|
11
|
Ghahremani PT, BaniArdalan S, Alehossein P, Parveen A, Jorjani M, Brown CM, Geldenhuys WJ, Huber JD, Ishrat T, Nasoohi S. Poststroke hyperglycemia dysregulates cap-dependent translation in neural cells. Life Sci 2025; 361:123336. [PMID: 39719167 DOI: 10.1016/j.lfs.2024.123336] [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: 08/06/2024] [Revised: 10/04/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
AIMS Post stroke hyperglycemia has been shown to deter functional recovery. Earlier findings have indicated the cap-dependent translation regulator 4E-BP1 is detrimentally upregulated in hyperglycemic conditions. The present study aims to test the hypothesis that hyperglycemic ischemic reperfusion injury (I/R) affects normal protein translation poststroke. METHODS Rat primary cortical neurons (PCNs) were exposed to oxygen glucose deprivation (OGD) followed by increasing glucose concentration (0, 5, 10, 25 mM) at reoxygenation. In vivo, adult rats were subjected to two hours transient distal middle cerebral artery occlusion (t-dMCAO) and hyperglycemic reperfusion. KEY FINDINGS In PCN cultures, high glucose levels impaired normal neurite growth at 24 h I/R where it drastically depressed S6 ribosomal protein phosphorylation at serine 235/236 residues in 40S ribosomal subunit. This concurred with substantial hypoxia inducible factor-1α (HIF-1α) destabilization and sustained vascular endothelial growth factor (VEGF). Our immunoblotting findings indicated HIF-1α stabilization and AMPK activation rely on glucose availability. Incremental glucose concentrations above the physiological levels, induced a shift towards 4E-BP1, eIF-4E hypo-phosphorylated forms leading to reduced eIF-4E availability and efficacy, as the key to recruit the 40S ribosomal subunit to the 5' end of mRNA. In vivo, immunostaining of t-dMCAO rat brains showed remarkable decrease in phosphorylated 4E-BP1 and particularly s6 ribosomal protein in the marginal cortical tissue of hyperglycemic compared to normoglycemic animals. SIGNIFICANCE These findings suggest a remarkable association between hyperglycemic I/R injury with dysregulated cap-dependent translation poststroke. Further loss/gain of function experiment may elucidate the potential therapeutic targets in regulation of HIF-1α/translation in hyperglycemic I/R injury.
Collapse
Affiliation(s)
| | - Soha BaniArdalan
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parsa Alehossein
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Arshi Parveen
- Department of Anatomy and Neurobiology, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Masoumeh Jorjani
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Candice M Brown
- Department of Neuroscience, School of Medicine, and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States
| | - Werner J Geldenhuys
- Department of Pharmaceutical Sciences, School of Pharmacy, Morgantown, WV, United States
| | - Jason D Huber
- Department of Pharmaceutical Sciences, School of Pharmacy, Morgantown, WV, United States
| | - Tauheed Ishrat
- Department of Anatomy and Neurobiology, School of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sanaz Nasoohi
- Neuroscience Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran; Department of Neuroscience, School of Medicine, and Rockefeller Neuroscience Institute, West Virginia University, Morgantown, WV, United States; Department of Pharmaceutical Sciences, School of Pharmacy, Morgantown, WV, United States; Department of Anesthesiology and Critical Care Medicine, School of Medicine, Johns Hopkins University, Baltimore, MD, United States.
| |
Collapse
|
12
|
Geng R, Wang Y, Wang R, Wu J, Bao X. Enhanced neurogenesis after ischemic stroke: the interplay between endogenous and exogenous stem cells. Neural Regen Res 2025; 21:01300535-990000000-00663. [PMID: 39820432 PMCID: PMC12094570 DOI: 10.4103/nrr.nrr-d-24-00879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 09/02/2024] [Accepted: 11/26/2024] [Indexed: 01/19/2025] Open
Abstract
ABSTRACT Ischemic stroke is a significant global health crisis, frequently resulting in disability or death, with limited therapeutic interventions available. Although various intrinsic reparative processes are initiated within the ischemic brain, these mechanisms are often insufficient to restore neuronal functionality. This has led to intensive investigation into the use of exogenous stem cells as a potential therapeutic option. This comprehensive review outlines the ontogeny and mechanisms of activation of endogenous neural stem cells within the adult brain following ischemic events, with focus on the impact of stem cell-based therapies on neural stem cells. Exogenous stem cells have been shown to enhance the proliferation of endogenous neural stem cells via direct cell-to-cell contact and through the secretion of growth factors and exosomes. Additionally, implanted stem cells may recruit host stem cells from their niches to the infarct area by establishing so-called "biobridges." Furthermore, xenogeneic and allogeneic stem cells can modify the microenvironment of the infarcted brain tissue through immunomodulatory and angiogenic effects, thereby supporting endogenous neuroregeneration. Given the convergence of regulatory pathways between exogenous and endogenous stem cells and the necessity for a supportive microenvironment, we discuss three strategies to simultaneously enhance the therapeutic efficacy of both cell types. These approaches include: (1) co-administration of various growth factors and pharmacological agents alongside stem cell transplantation to reduce stem cell apoptosis; (2) synergistic administration of stem cells and their exosomes to amplify paracrine effects; and (3) integration of stem cells within hydrogels, which provide a protective scaffold for the implanted cells while facilitating the regeneration of neural tissue and the reconstitution of neural circuits. This comprehensive review highlights the interactions and shared regulatory mechanisms between endogenous neural stem cells and exogenously implanted stem cells and may offer new insights for improving the efficacy of stem cell-based therapies in the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Ruxu Geng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Yuhe Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jun Wu
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- State Key Laboratory of Common Mechanism Research for Major Diseases, Beijing, China
| |
Collapse
|
13
|
Kim S, Jung UJ, Kim SR. The Crucial Role of the Blood-Brain Barrier in Neurodegenerative Diseases: Mechanisms of Disruption and Therapeutic Implications. J Clin Med 2025; 14:386. [PMID: 39860392 PMCID: PMC11765772 DOI: 10.3390/jcm14020386] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
The blood-brain barrier (BBB) is a crucial structure that maintains brain homeostasis by regulating the entry of molecules and cells from the bloodstream into the central nervous system (CNS). Neurodegenerative diseases such as Alzheimer's and Parkinson's disease, as well as ischemic stroke, compromise the integrity of the BBB. This leads to increased permeability and the infiltration of harmful substances, thereby accelerating neurodegeneration. In this review, we explore the mechanisms underlying BBB disruption, including oxidative stress, neuroinflammation, vascular dysfunction, and the loss of tight junction integrity, in patients with neurodegenerative diseases. We discuss how BBB breakdown contributes to neuroinflammation, neurotoxicity, and the abnormal accumulation of pathological proteins, all of which exacerbate neuronal damage and facilitate disease progression. Furthermore, we discuss potential therapeutic strategies aimed at preserving or restoring BBB function, such as anti-inflammatory treatments, antioxidant therapies, and approaches to enhance tight junction integrity. Given the central role of the BBB in neurodegeneration, maintaining its integrity represents a promising therapeutic approach to slow or prevent the progression of neurodegenerative diseases.
Collapse
Affiliation(s)
- Sehwan Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Un Ju Jung
- Department of Food Science and Nutrition, Pukyong National University, Busan 48513, Republic of Korea;
| | - Sang Ryong Kim
- School of Life Science and Biotechnology, Kyungpook National University, Daegu 41566, Republic of Korea;
- BK21 FOUR KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Republic of Korea
- Brain Science and Engineering Institute, Kyungpook National University, Daegu 41404, Republic of Korea
| |
Collapse
|
14
|
Niu B, Wu H, Li Y, Klugah-Brown B, Hanna G, Yao Y, Jing J, Baig TI, Xia Y, Yao D, Biswal B. Topological functional network analysis of cortical blood flow in hyperacute ischemic rats. Brain Struct Funct 2024; 230:20. [PMID: 39724244 PMCID: PMC11671571 DOI: 10.1007/s00429-024-02864-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 11/29/2024] [Indexed: 12/28/2024]
Abstract
Acute cerebral ischemia alters brain network connectivity, leading to notable increases in both anatomical and functional connectivity while observing a reduction in metabolic connectivity. However, alterations of the cerebral blood flow (CBF) based functional connectivity remain unclear. We collected continuous CBF images using laser speckle contrast imaging (LSCI) technology to monitor ischemic occlusion-reperfusion progression through occlusion of the left carotid artery. We also used a dense cortical grid atlas to construct CBF-based functional connectivity networks for hyperacute ischemic rodents. Graph theoretical analysis was used to measure network topological characteristics and construct topological connection graphs. Coactivation pattern (CAP) analysis was utilized to examine the spatiotemporal characteristics of the global network. Additionally, we measured evoked functional hyperemia and correlated it with network topologies. Network analysis indicated a significant increase in functional connectivity, global efficiency, local efficiency, small-worldness, clustering coefficient, and regional degree centrality primarily within the left ischemic intra-hemisphere, accompanied by weaker changes in the right intra-hemisphere. Inter-hemisphere networks exhibited reduced homologous connections, global efficiency, and small-worldness. CAP analysis revealed increased strength of the left negative activation brain network's state fraction of time and transition probability from equilibrium-to-imbalance states. Left network metrics declined following blood flow reperfusion. Furthermore, positive/negative correlations between barrel-evoked intensity and regional network topologies were reversed as negative/positive correlations after cerebral ischemia. These findings suggest a damaged CBF functional network mechanism following acute cerebral ischemia and a disrupted association between resting state and evoked hyperemia.
Collapse
Affiliation(s)
- Bochao Niu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Hongzhou Wu
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yilu Li
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Benjamin Klugah-Brown
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - George Hanna
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA
| | - Youwang Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Junlin Jing
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Talha Imtiaz Baig
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Yang Xia
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Dezhong Yao
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Bharat Biswal
- The Clinical Hospital of Chengdu Brain Science Institute, MOE Key Laboratory for Neuroinformation, School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, 611731, China.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ, 07102, USA.
| |
Collapse
|
15
|
Ball BK, Hyun Park J, Proctor EA, Brubaker DK. Cross-disease modeling of peripheral blood identifies biomarkers of type 2 diabetes predictive of Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.11.627991. [PMID: 39713369 PMCID: PMC11661382 DOI: 10.1101/2024.12.11.627991] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Type 2 diabetes (T2D) is a significant risk factor for Alzheimer's disease (AD). Despite multiple studies reporting this connection, the mechanism by which T2D exacerbates AD is poorly understood. It is challenging to design studies that address co-occurring and comorbid diseases, limiting the number of existing evidence bases. To address this challenge, we expanded the applications of a computational framework called Translatable Components Regression (TransComp-R), initially designed for cross-species translation modeling, to perform cross-disease modeling to identify biological programs of T2D that may exacerbate AD pathology. Using TransComp-R, we combined peripheral blood-derived T2D and AD human transcriptomic data to identify T2D principal components predictive of AD status. Our model revealed genes enriched for biological pathways associated with inflammation, metabolism, and signaling pathways from T2D principal components predictive of AD. The same T2D PC predictive of AD outcomes unveiled sex-based differences across the AD datasets. We performed a gene expression correlational analysis to identify therapeutic hypotheses tailored to the T2D-AD axis. We identified six T2D and two dementia medications that induced gene expression profiles associated with a non-T2D or non-AD state. Finally, we assessed our blood-based T2DxAD biomarker signature in post-mortem human AD and control brain gene expression data from the hippocampus, entorhinal cortex, superior frontal gyrus, and postcentral gyrus. Using partial least squares discriminant analysis, we identified a subset of genes from our cross-disease blood-based biomarker panel that significantly separated AD and control brain samples. Our methodological advance in cross-disease modeling identified biological programs in T2D that may predict the future onset of AD in this population. This, paired with our therapeutic gene expression correlational analysis, also revealed alogliptin, a T2D medication that may help prevent the onset of AD in T2D patients.
Collapse
Affiliation(s)
- Brendan K. Ball
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Jee Hyun Park
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Elizabeth A. Proctor
- Department of Neurosurgery, Penn State College of Medicine, Hershey, PA, USA
- Department of Pharmacology, Penn State College of Medicine, Hershey, PA, USA
- Department of Biomedical Engineering, Penn State University, State College, PA, USA
- Center for Neural Engineering, Penn State University, State College, PA, USA
- Department of Engineering Science & Mechanics, Penn State University, State College, PA, USA
| | - Douglas K. Brubaker
- Center for Global Health & Diseases, Department of Pathology, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Blood Heart Lung Immunology Research Center, University Hospitals, Cleveland, OH, USA
| |
Collapse
|
16
|
Leng C, Lin K, Zhou M, Tao X, Sun B, Shu X, Liu W. Apolipoprotein E deficiency exacerbates blood-brain barrier disruption and hyperglycemia-associated hemorrhagic transformation after ischemic stroke. J Stroke Cerebrovasc Dis 2024; 33:107987. [PMID: 39218418 DOI: 10.1016/j.jstrokecerebrovasdis.2024.107987] [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: 04/28/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The polymorphism of the apolipoprotein E (ApoE) gene has been implicated in both the susceptibility to neurodegenerative disease and the prognosis of traumatic brain injury (TBI). However, the influence of ApoE on the risk of hemorrhagic transformation (HT) after acute ischemic stroke remains inconclusive. The present study aimed to investigate the potential impact of ApoE deficiency on the risk of hyperglycemia-associated HT and to elucidate the underlying mechanisms. METHODS Wild-type (WT) and ApoE knockout (ApoE-/-) mice were injected with 50 % glucose to induce hyperglycemia and subsequently subjected to 90 min of intraluminal middle cerebral artery occlusion (MCAO). The mortality, neurological function, HT incidence and HT grading-score were evaluated at 24 hours after reperfusion. To evaluate the integrity of blood-brain barrier (BBB), the immunoglobulin G (IgG) leakage and the protein expressions of tight junctions (TJs) were detected using immunofluorescent staining and western blotting. Finally, the levels of matrix metalloproteinases (MMP)-2/9, microglial activation and proinflammatory mediators were investigated using immunofluorescent staining and western blotting. RESULTS ApoE-/- mice exhibited increased mortality and exacerbated neurological impairment, concomitant with more severe hyperglycemia-associated HT 24 hours post-reperfusion. Meanwhile, ApoE deficiency exacerbated the disruption of BBB, characterized by increased leakage of IgG, aggravated degradation of TJs and microvascular basement membranes. Furthermore, ApoE deficiency further aggravated the upregulation of MMP-2/9 and microglia-triggered neuroinflammation. CONCLUSIONS Our findings demonstrate that the absence of ApoE exacerbates neurological impairment and hyperglycemia-associated HT in ischemic stroke mice, which is closely associated with MMP-2/9 signaling and neuroinflammation-mediated disruption of BBB.
Collapse
Affiliation(s)
- Changlong Leng
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China.
| | - Kuan Lin
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China.
| | - Mei Zhou
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China.
| | - Xiaoqin Tao
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China.
| | - Binlian Sun
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China.
| | - Xiji Shu
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China.
| | - Wei Liu
- Hubei Key Laboratory of Cognitive and Affective Disorder, Jianghan University, Wuhan, China; Institute of Cerebrovascular Disease, School of Medicine, Jianghan University, Wuhan, China; Department of Pathology and Pathophysiology, School of Medicine, Jianghan University, Wuhan, China.
| |
Collapse
|
17
|
Daga KR, Larey AM, Morfin MG, Chen K, Bitarafan S, Carpenter JM, Hynds HM, Hines KM, Wood LB, Marklein RA. Microglia morphological response to mesenchymal stromal cell extracellular vesicles demonstrates EV therapeutic potential for modulating neuroinflammation. J Biol Eng 2024; 18:58. [PMID: 39420399 PMCID: PMC11488223 DOI: 10.1186/s13036-024-00449-w] [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: 06/26/2024] [Accepted: 09/16/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND Mesenchymal stromal cell derived extracellular vesicles (MSC-EVs) are a promising therapeutic for neuroinflammation. MSC-EVs can interact with microglia, the resident immune cells of the brain, to exert their immunomodulatory effects. In response to inflammatory cues, such as cytokines, microglia undergo phenotypic changes indicative of their function e.g. morphology and secretion. However, these changes in response to MSC-EVs are not well understood. Additionally, no disease-relevant screening tools to assess MSC-EV bioactivity exist, which has further impeded clinical translation. Here, we developed a quantitative, high throughput morphological profiling approach to assess the response of microglia to neuroinflammation- relevant signals and whether this morphological response can be used to indicate the bioactivity of MSC-EVs. RESULTS Using an immortalized human microglia cell-line, we observed increased size (perimeter, major axis length) and complexity (form factor) upon stimulation with interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Upon treatment with MSC-EVs, the overall morphological score (determined using principal component analysis) shifted towards the unstimulated morphology, indicating that MSC-EVs are bioactive and modulate microglia. The morphological effects of MSC-EVs in TNF-α /IFN-γ stimulated cells were concomitant with reduced secretion of 14 chemokines/cytokines (e.g. CXCL6, CXCL9) and increased secretion of 12 chemokines/cytokines (e.g. CXCL8, CXCL10). Proteomic analysis of cell lysates revealed significant increases in 192 proteins (e.g. HIBADH, MEAK7, LAMC1) and decreases in 257 proteins (e.g. PTEN, TOM1, MFF) with MSC-EV treatment. Of note, many of these proteins are involved in regulation of cell morphology and migration. Gene Set Variation Analysis revealed upregulation of pathways associated with immune response, such as regulation of cytokine production, immune cell infiltration (e.g. T cells, NK cells) and morphological changes (e.g. Semaphorin, RHO/Rac signaling). Additionally, changes in microglia mitochondrial morphology were measured suggesting that MSC-EV modulate mitochondrial metabolism. CONCLUSION This study comprehensively demonstrates the effects of MSC-EVs on human microglial morphology, cytokine secretion, cellular proteome, and mitochondrial content. Our high-throughput, rapid, low-cost morphometric approach enables screening of MSC-EV batches and manufacturing conditions to enhance EV function and mitigate EV functional heterogeneity in a disease relevant manner. This approach is highly generalizable and can be further adapted and refined based on selection of the disease-relevant signal, target cell, and therapeutic product.
Collapse
Affiliation(s)
- Kanupriya R Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Andrew M Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G Morfin
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
| | - Kailin Chen
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Sara Bitarafan
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Hannah M Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ross A Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA.
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA.
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, 10903 New Hampshire Ave, Silver Spring, MD, 20903, USA.
| |
Collapse
|
18
|
Xie L, Wu Q, Li K, Khan MAS, Zhang A, Sinha B, Li S, Chang SL, Brody DL, Grinstaff MW, Zhou S, Alterovitz G, Liu P, Wang X. Tryptophan Metabolism in Alzheimer's Disease with the Involvement of Microglia and Astrocyte Crosstalk and Gut-Brain Axis. Aging Dis 2024; 15:2168-2190. [PMID: 38916729 PMCID: PMC11346405 DOI: 10.14336/ad.2024.0134] [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: 02/02/2024] [Accepted: 05/03/2024] [Indexed: 06/26/2024] Open
Abstract
Alzheimer's disease (AD) is an age-dependent neurodegenerative disease characterized by extracellular Amyloid Aβ peptide (Aβ) deposition and intracellular Tau protein aggregation. Glia, especially microglia and astrocytes are core participants during the progression of AD and these cells are the mediators of Aβ clearance and degradation. The microbiota-gut-brain axis (MGBA) is a complex interactive network between the gut and brain involved in neurodegeneration. MGBA affects the function of glia in the central nervous system (CNS), and microbial metabolites regulate the communication between astrocytes and microglia; however, whether such communication is part of AD pathophysiology remains unknown. One of the potential links in bilateral gut-brain communication is tryptophan (Trp) metabolism. The microbiota-originated Trp and its metabolites enter the CNS to control microglial activation, and the activated microglia subsequently affect astrocyte functions. The present review highlights the role of MGBA in AD pathology, especially the roles of Trp per se and its metabolism as a part of the gut microbiota and brain communications. We (i) discuss the roles of Trp derivatives in microglia-astrocyte crosstalk from a bioinformatics perspective, (ii) describe the role of glia polarization in the microglia-astrocyte crosstalk and AD pathology, and (iii) summarize the potential of Trp metabolism as a therapeutic target. Finally, we review the role of Trp in AD from the perspective of the gut-brain axis and microglia, as well as astrocyte crosstalk, to inspire the discovery of novel AD therapeutics.
Collapse
Affiliation(s)
- Lushuang Xie
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Qiaofeng Wu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Kelin Li
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Mohammed A. S. Khan
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Andrew Zhang
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Bharati Sinha
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Sihui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, 610075, China.
| | - Sulie L. Chang
- Department of Biological Sciences, Institute of NeuroImmune Pharmacology, Seton Hall University, South Orange, NJ 07079, USA.
| | - David L. Brody
- Department of Neurology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA.
| | | | - Shuanhu Zhou
- Harvard Medical School, Harvard Stem Cell Institute, Boston, MA 02115, USA.
| | - Gil Alterovitz
- Biomedical Cybernetics Laboratory, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | - Pinghua Liu
- Department of Chemistry, Boston University, Boston, MA 02215, USA.
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Ni G, Kou L, Duan C, Meng R, Wang P. MicroRNA-199a-5p attenuates blood-brain barrier disruption following ischemic stroke by regulating PI3K/Akt signaling pathway. PLoS One 2024; 19:e0306793. [PMID: 39302945 DOI: 10.1371/journal.pone.0306793] [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: 02/01/2024] [Accepted: 06/24/2024] [Indexed: 09/22/2024] Open
Abstract
OBJECTIVE To explore whether miR-199a-5p regulated BBB integrity through PI3K/Akt pathway after ischemia stroke. METHODS Adult male Sprague-Dawley rats with permanent middle cerebral artery occlusion(MCAO) were used in experiment. The Ludmila Belayev 12-point scoring was used to measure the neurological function of MCAO rats. The Evans Blue Stain, immunofluorescence staining, western-blotting and RT-PCR were performed to evaluate the effects of miR-199a-5p mimic on BBB integrity in rats following MCAO. RESULTS The result suggested that miR-199a-5p mimic treatment possessed the potential to boost proprioception and motor activity of MCAO rats. MiR-199a-5p decreased the expression of PIK3R2 after MCAO, activated Akt signaling pathway, and increased the expression of Claudin-5 and VEGF in the ischemic penumbra. Furthermore, miR-199a-5p alleviated inflammation after cerebral ischemia. BBB leakage and neurocyte apoptosis were cut down in MCAO rats treated with miR-199a-5p mimic. CONCLUSIONS MiR-199a-5p mimic decreased the expression of PIK3R2 and activated Akt signaling pathway after ischemia stroke, reduced the expression of inflammatory cytokines, and attenuated BBB disruption after ischemic stroke.
Collapse
Affiliation(s)
- Guangxiao Ni
- Department of Rehabilitation of the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Lulu Kou
- Department of Rehabilitation of the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Chunqiao Duan
- Department of Rehabilitation of the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Ran Meng
- Department of Rehabilitation of the Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Pu Wang
- Stomatological Laboratory of the Second Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
20
|
Liu S, Bai T, Feng J. Endocan, a novel glycoprotein with multiple biological activities, may play important roles in neurological diseases. Front Aging Neurosci 2024; 16:1438367. [PMID: 39328246 PMCID: PMC11426085 DOI: 10.3389/fnagi.2024.1438367] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Endothelial cell specific-1 (ESM-1), also known as endocan, is a soluble dermatan sulfate proteoglycan that is mainly secreted by endothelial cells. Endocan is associated with tumorigenesis and cancer progression and is also related to cardiovascular disorders, autoimmune diseases, and sepsis. The phenylalanine-rich region and linear polysaccharide of endocan are necessary for the protein to exert its biological functions. Elevated plasma endocan levels reflect endothelial activation and dysfunction. In addition, endocan participates in complex inflammatory responses and proliferative processes. Here, we reviewed current research on endocan, elaborated the protein's structure and biological functions, and speculated on its possible clinical value in nervous system diseases. We conclude that endocan may be a glycoprotein that plays an important role in neurological disorders.
Collapse
Affiliation(s)
- Shuo Liu
- The Fourth People's Hospital of Shenyang, Shenyang, Liaoning, China
- Department of Neurology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| | - Tao Bai
- Department of Neurology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| | - Juan Feng
- Department of Neurology, Shengjing Hospital of China Medical University, China Medical University, Shenyang, China
| |
Collapse
|
21
|
Li ZR, Wang YY, Wang ZH, Qin QL, Huang C, Shi GS, He HY, Deng YH, He XY, Zhao XM. The positive role of transforming growth factor-β1 in ischemic stroke. Cell Signal 2024; 121:111301. [PMID: 39019338 DOI: 10.1016/j.cellsig.2024.111301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 07/05/2024] [Accepted: 07/12/2024] [Indexed: 07/19/2024]
Abstract
Ischemic stroke is one of the most disabling and fatal diseases around the world. The damaged brain tissues will undergo excessive autophagy, vascular endothelial cells injury, blood-brain barrier (BBB) impairment and neuroinflammation after ischemic stroke. However, there is no unified viewpoint on the underlying mechanism of brain damage. Transforming growth factor-β1 (TGF-β1), as a multi-functional cytokine, plays a crucial role in the intricate pathological processes and helps maintain the physiological homeostasis of brain tissues through various signaling pathways after ischemic stroke. In this review, we summarize the protective role of TGF-β1 in autophagic flux, BBB, vascular remodeling, neuroinflammation and other aspects after ischemic stroke. Based on the review, we believe that TGF-β1 could serve as a key target for treating ischemic stroke.
Collapse
Affiliation(s)
- Zi-Rong Li
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Yong-Yan Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Zi-Han Wang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Qi-Lin Qin
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Cheng Huang
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Guang-Sen Shi
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Hong-Yun He
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| | - Yi-Hao Deng
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China.
| | - Xiu-Ying He
- Department of Anesthesiology, Institute of Neurological Disease, West China Hospital, Sichuan University, Chengdu, China.
| | - Xiao-Ming Zhao
- Faculty of Medicine, Kunming University of Science and Technology, Kunming, China; Anning First People's Hospital Affiliated to Kunming University of Science and Technology, Kunming, China.
| |
Collapse
|
22
|
Hu Y, Huang S, Shen T, Wang R, Geng M, Wang Y, Zheng Y, Luo Y, Li S. Prognostic Significance of Plasma VEGFA and VEGFR2 in Acute Ischemic Stroke-a Prospective Cohort Study. Mol Neurobiol 2024; 61:6341-6353. [PMID: 38300447 DOI: 10.1007/s12035-024-03973-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 01/12/2024] [Indexed: 02/02/2024]
Abstract
Enhancement of vascular remodeling in affected brain tissue is a novel therapy for acute ischemic stroke (AIS). However, conclusions regarding angiogenesis after AIS remain ambiguous. Vascular endothelial growth factor A (VEGFA) and VEGF receptor 2 (VEGFR2) are potent regulators of angiogenesis and vascular permeability. We aimed to investigate the association between VEGFA/VEGFR2 expression in the acute stage of stroke and prognosis of patients with AIS. We enrolled 120 patients with AIS within 24 h of stroke onset and 26 healthy controls. Plasma levels of VEGFA and VEGFR2 were measured by enzyme-linked immunosorbent assay (ELISA). The primary endpoint was an unfavorable outcome defined as a modified Rankin Scale (mRS) score > 2 at 3 months after AIS. Univariate and multivariate logistic regression analyses were used to identify risk factors affecting prognosis. Plasma VEGFA and VEGFR2 were significantly higher in patients with AIS than in health controls, and also significantly higher in patients with unfavorable than those with favorable outcomes. Moreover, both VEGFA and VEGFR2 showed a significantly positive correlation with mRS at 3 months. Univariate and multivariate analyses showed VEGFA and VEGFR2 remained associated with unfavorable outcomes, and adding VEGFA and VEGFR2 to the clinical model significantly improved risk reclassification (continuous net reclassification improvement, 105.71%; integrated discrimination improvement, 23.45%). The new risk model curve exhibited a good fit with an area under the receiver operating characteristic curve (ROC) curve of 0.9166 (0.8658-0.9674). Plasma VEGFA and VEGFR2 are potential markers for predicting prognosis; thus these two plasma biomarkers may improve risk stratification in patients with AIS.
Collapse
Affiliation(s)
- Yue Hu
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Shuangfeng Huang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Tong Shen
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Rongliang Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Meng Geng
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yilin Wang
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yangmin Zheng
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yumin Luo
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| | - Sijie Li
- Institute of Cerebrovascular Disease Research and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
23
|
Hu B, Pei J, Wan C, Liu S, Xu Z, Zou Y, Li Z, Tang Z. Mechanisms of Postischemic Stroke Angiogenesis: A Multifaceted Approach. J Inflamm Res 2024; 17:4625-4646. [PMID: 39045531 PMCID: PMC11264385 DOI: 10.2147/jir.s461427] [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: 02/22/2024] [Accepted: 06/26/2024] [Indexed: 07/25/2024] Open
Abstract
Ischemic stroke constitutes a significant global health care challenge, and a comprehensive understanding of its recovery mechanisms is imperative for the development of innovative therapeutic strategies. Angiogenesis, a pivotal element of ischemic tissue repair, facilitates the restoration of blood flow to damaged regions, thereby promoting neuronal regeneration and functional recovery. Nevertheless, the mechanisms underlying postischemic stroke angiogenesis remain incompletely elucidated. This review meticulously examines the constituents of the neurovascular unit, ion channels, molecular mediators, and signaling pathways implicated in angiogenesis following stroke. Furthermore, it delves into prospective therapeutic strategies informed by these factors. Our objective is to provide detailed and exhaustive information on the intricate mechanisms governing postischemic stroke angiogenesis, thus providing a robust scientific foundation for the advancement of novel neurorepair therapies.
Collapse
Affiliation(s)
- Bin Hu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Jingchun Pei
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Cheng Wan
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
- Department of Medical Imaging, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Shuangshuang Liu
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhe Xu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Kunming Medical University, Kunming, People’s Republic of China
- School of Basic Medical Sciences, Qujing Medical College, Qujing, People’s Republic of China
| | - Yongwei Zou
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhigao Li
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| | - Zhiwei Tang
- Department of Neurosurgery, The First Affiliated Hospital of Kunming Medical University, Kunming, People’s Republic of China
| |
Collapse
|
24
|
Cha Z, Qiao Y, Lu Q, Wang Q, Lu X, Zhou H, Li T. Research progress and challenges of stem cell therapy for ischemic stroke. Front Cell Dev Biol 2024; 12:1410732. [PMID: 39040041 PMCID: PMC11260720 DOI: 10.3389/fcell.2024.1410732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 06/17/2024] [Indexed: 07/24/2024] Open
Abstract
Ischemic stroke is a significant global cause of death and disability. Currently, treatment options for acute ischemic stroke are limited to intravenous thrombolysis and mechanical recanalization. Therefore, novel neuroprotective strategies are imperative. Stem cell transplantation possesses the capabilities of differentiation, proliferation, neuronal replacement, nerve pathway reconstruction, secretion of nerve growth factors, and enhancement of the microenvironment; thus, it is a potential therapeutic approach for ischemic stroke. In addition, the immunomodulatory function of stem cells and the combined treatment of stem cells and exosomes exhibit a favorable protective effect on brain injury and neurological dysfunction following stroke. Meanwhile, the theory of microbiota-gut-brain axis provides us with a novel perspective for comprehending and managing neurological diseases. Lastly, stem cell transplantation has demonstrated promising outcomes not only in treating ischemic stroke but also in dealing with other neurological disorders, such as brain tumors. Furthermore, challenges related to the tissue source, delivery method, immune response, and timing of transplantation still need to be addressed to optimize the treatment.
Collapse
Affiliation(s)
- Zaihong Cha
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yisheng Qiao
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qixiong Lu
- The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Qiyang Wang
- Department of Orthopedics, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiaoyang Lu
- Department of Neurosurgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Hu Zhou
- Department of Neurosurgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Tao Li
- Research Center for Clinical Medicine, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Institute of Neurosurgery and Neuroscience, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| |
Collapse
|
25
|
Daga KR, Larey AM, Morfin MG, Chen K, Bitarafan S, Carpenter JM, Hynds HM, Hines KM, Wood LB, Marklein RA. Microglia Morphological Response to Mesenchymal Stromal Cell Extracellular Vesicles Demonstrates EV Therapeutic Potential for Modulating Neuroinflammation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.01.601612. [PMID: 39005342 PMCID: PMC11245023 DOI: 10.1101/2024.07.01.601612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Background Mesenchymal stromal cell derived extracellular vesicles (MSC-EVs) are a promising therapeutic for neuroinflammation. MSC-EVs can interact with microglia, the resident immune cells of the brain, to exert their immunomodulatory effects. In response to inflammatory cues, such as cytokines, microglia undergo phenotypic changes indicative of their function e.g. morphology and secretion. However, these changes in response to MSC-EVs are not well understood. Additionally, no disease-relevant screening tools to assess MSC-EV bioactivity exist, which has further impeded clinical translation. Here, we developed a quantitative, high throughput morphological profiling approach to assess the response of microglia to neuroinflammation-relevant signals and whether this morphological response can be used to indicate the bioactivity of MSC-EVs. Results Using an immortalized human microglia cell-line, we observed increased size (perimeter, major axis length) and complexity (form factor) upon stimulation with interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α). Upon treatment with MSC-EVs, the overall morphological score (determined using principal component analysis) shifted towards the unstimulated morphology, indicating that MSC-EVs are bioactive and modulate microglia. The morphological effects of MSC-EVs in TNF-γ/IFN-α stimulated cells were concomitant with reduced secretion of 14 chemokines/cytokines (e.g. CXCL6, CXCL9) and increased secretion of 12 chemokines/cytokines (e.g. CXCL8, CXCL10). Proteomic analysis of cell lysates revealed significant increases in 192 proteins (e.g. HIBADH, MEAK7, LAMC1) and decreases in 257 proteins (e.g. PTEN, TOM1, MFF) with MSC-EV treatment. Of note, many of these proteins are involved in regulation of cell morphology and migration. Gene Set Variation Analysis revealed upregulation of pathways associated with immune response, such as regulation of cytokine production, immune cell infiltration (e.g. T cells, NK cells) and morphological changes (e.g. Semaphorin, RHO/Rac signaling). Additionally, changes in microglia mitochondrial morphology were measured suggesting that MSC-EV modulate mitochondrial metabolism. Conclusion This study comprehensively demonstrates the effects of MSC-EVs on human microglial morphology, cytokine secretion, cellular proteome, and mitochondrial content. Our high-throughput, rapid, low-cost morphological approach enables screening of MSC-EV batches and manufacturing conditions to enhance EV function and mitigate EV functional heterogeneity in a disease relevant manner. This approach is highly generalizable and can be further adapted and refined based on selection of the disease-relevant signal, target cell, and therapeutic product.
Collapse
Affiliation(s)
- Kanupriya R Daga
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Andrew M Larey
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
| | - Maria G Morfin
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
| | - Kailin Chen
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Franklin College of Arts and Sciences, University of Georgia, Athens, GA, USA
| | - Sara Bitarafan
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Hannah M Hynds
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Kelly M Hines
- Department of Chemistry, University of Georgia, Athens, GA, USA
| | - Levi B Wood
- George W. Woodruff School of Mechanical Engineering and Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ross A Marklein
- School of Chemical, Materials, and Biomedical Engineering, University of Georgia, Athens, GA, USA
- Regenerative Bioscience Center, University of Georgia, Athens, GA, USA
- Center for Biologics Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
26
|
Liu C, Guo Y, Deng S, Zhou S, Wu S, Chen T, Shi X, Mamtilahun M, Xu T, Liu Z, Li H, Zhang Z, Tian H, Chung WS, Wang J, Yang GY, Tang Y. Hemorrhagic stroke-induced subtype of inflammatory reactive astrocytes disrupts blood-brain barrier. J Cereb Blood Flow Metab 2024; 44:1102-1116. [PMID: 38388375 PMCID: PMC11179611 DOI: 10.1177/0271678x241235008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/17/2023] [Accepted: 02/06/2024] [Indexed: 02/24/2024]
Abstract
Astrocytes undergo disease-specific transcriptomic changes upon brain injury. However, phenotypic changes of astrocytes and their functions remain unclear after hemorrhagic stroke. Here we reported hemorrhagic stroke induced a group of inflammatory reactive astrocytes with high expression of Gfap and Vimentin, as well as inflammation-related genes lipocalin-2 (Lcn2), Complement component 3 (C3), and Serpina3n. In addition, we demonstrated that depletion of microglia but not macrophages inhibited the expression of inflammation-related genes in inflammatory reactive astrocytes. RNA sequencing showed that blood-brain barrier (BBB) disruption-related gene matrix metalloproteinase-3 (MMP3) was highly upregulated in inflammatory reactive astrocytes. Pharmacological inhibition of MMP3 in astrocytes or specific deletion of astrocytic MMP3 reduced BBB disruption and improved neurological outcomes of hemorrhagic stroke mice. Our study demonstrated that hemorrhagic stroke induced a group of inflammatory reactive astrocytes that were actively involved in disrupting BBB through MMP3, highlighting a specific group of inflammatory reactive astrocytes as a critical driver for BBB disruption in neurological diseases.
Collapse
Affiliation(s)
- Chang Liu
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yiyan Guo
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyu Deng
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shiyi Zhou
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Shengju Wu
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Chen
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojing Shi
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Muyassar Mamtilahun
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Tongtong Xu
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Ze Liu
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hanlai Li
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Zhijun Zhang
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Hengli Tian
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Won-Suk Chung
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jixian Wang
- Department of Rehabilitation Medicine, Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Guo-Yuan Yang
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| | - Yaohui Tang
- Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
27
|
Amin N, Abbasi IN, Wu F, Shi Z, Sundus J, Badry A, Yuan X, Zhao BX, Pan J, Mi XD, Luo Y, Geng Y, Fang M. The Janus face of HIF-1α in ischemic stroke and the possible associated pathways. Neurochem Int 2024; 177:105747. [PMID: 38657682 DOI: 10.1016/j.neuint.2024.105747] [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: 10/30/2023] [Revised: 03/01/2024] [Accepted: 04/19/2024] [Indexed: 04/26/2024]
Abstract
Stroke is the most devastating disease, causing paralysis and eventually death. Many clinical and experimental trials have been done in search of a new safe and efficient medicine; nevertheless, scientists have yet to discover successful remedies that are also free of adverse effects. This is owing to the variability in intensity, localization, medication routes, and each patient's immune system reaction. HIF-1α represents the modern tool employed to treat stroke diseases due to its functions: downstream genes such as glucose metabolism, angiogenesis, erythropoiesis, and cell survival. Its role can be achieved via two downstream EPO and VEGF strongly related to apoptosis and antioxidant processes. Recently, scientists paid more attention to drugs dealing with the HIF-1 pathway. This review focuses on medicines used for ischemia treatment and their potential HIF-1α pathways. Furthermore, we discussed the interaction between HIF-1α and other biological pathways such as oxidative stress; however, a spotlight has been focused on certain potential signalling contributed to the HIF-1α pathway. HIF-1α is an essential regulator of oxygen balance within cells which affects and controls the expression of thousands of genes related to sustaining homeostasis as oxygen levels fluctuate. HIF-1α's role in ischemic stroke strongly depends on the duration and severity of brain damage after onset. HIF-1α remains difficult to investigate, particularly in ischemic stroke, due to alterations in the acute and chronic phases of the disease, as well as discrepancies between the penumbra and ischemic core. This review emphasizes these contrasts and analyzes the future of this intriguing and demanding field.
Collapse
Affiliation(s)
- Nashwa Amin
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China; Department of Zoology, Faculty of Science, Aswan University, Egypt; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Irum Naz Abbasi
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Wu
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Zongjie Shi
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Javaria Sundus
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Azhar Badry
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Xia Yuan
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China
| | - Bing-Xin Zhao
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jie Pan
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Xiao-Dan Mi
- Center for Rehabilitation Medicine, Rehabilitation & Sports Medicine Research Institute of Zhejiang Province, Department of Rehabilitation Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yuhuan Luo
- Department of Pediatrics, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Geng
- Center for Rehabilitation Medicine, Department of Neurology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Marong Fang
- Institute of Systemic Medicine, Zhejiang University School of Medicine, Hangzhou, China; Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| |
Collapse
|
28
|
Wang X, Li A, Fan H, Li Y, Yang N, Tang Y. Astrocyte-Derived Extracellular Vesicles for Ischemic Stroke: Therapeutic Potential and Prospective. Aging Dis 2024; 15:1227-1254. [PMID: 37728588 PMCID: PMC11081164 DOI: 10.14336/ad.2023.0823-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 08/23/2023] [Indexed: 09/21/2023] Open
Abstract
Stroke is a leading cause of death and disability in the world. Astrocytes are special glial cells within the central nervous system and play important roles in mediating neuroprotection and repair processes during stroke. Extracellular vesicles (EVs) are lipid bilayer particles released from cells that facilitate intercellular communication in stroke by delivering proteins, lipids, and RNA to target cells. Recently, accumulating evidence suggested that astrocyte-derived EVs (ADEVs) are actively involved in mediating numerous biological processes including neuroprotection and neurorepair in stroke and they are realized as an excellent therapeutic approach for treating stroke. In this review we systematically summarize the up-to-date research on ADEVs in stroke, and prospects for its potential as a novel therapeutic target for stroke. We also provide an overview of the effects and functions of ADEVs on stroke recovery, which may lead to developing clinically relevant therapies for stroke.
Collapse
Affiliation(s)
- Xianghui Wang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Aihua Li
- Department of rehabilitation medicine, Jinan Hospital, Jinan, China
| | - Huaju Fan
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
| | - Yanyan Li
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
| | - Nana Yang
- School of Bioscience and Technology, Weifang Medical University, Weifang, Shandong, China.
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | - Yaohui Tang
- School of Biomedical Engineering and Affiliated Sixth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
29
|
Sakamuri SSVP, Sure VN, Oruganti L, Wisen W, Chandra PK, Liu N, Fonseca VA, Wang X, Klein J, Katakam PVG. Acute severe hypoglycemia alters mouse brain microvascular proteome. J Cereb Blood Flow Metab 2024; 44:556-572. [PMID: 37944245 PMCID: PMC10981402 DOI: 10.1177/0271678x231212961] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 09/12/2023] [Accepted: 10/05/2023] [Indexed: 11/12/2023]
Abstract
Hypoglycemia increases the risk related to stroke and neurodegenerative diseases, however, the underlying mechanisms are unclear. For the first time, we studied the effect of a single episode (acute) of severe (ASH) and mild (AMH) hypoglycemia on mouse brain microvascular proteome. After four-hour fasting, insulin was administered (i.p) to lower mean blood glucose in mice and induce ∼30 minutes of ASH (∼30 mg/dL) or AMH (∼75 mg/dL), whereas a similar volume of saline was given to control mice (∼130 mg/dL). Blood glucose was allowed to recover over 60 minutes either spontaneously or by 20% dextrose administration (i.p). Twenty-four hours later, the brain microvessels (BMVs) were isolated, and tandem mass tag (TMT)-based quantitative proteomics was performed using liquid chromatography-mass spectrometry (LC/MS). When compared to control, ASH significantly downregulated 13 proteins (p ≤ 0.05) whereas 23 proteins showed a strong trend toward decrease (p ≤ 0.10). When compared to AMH, ASH significantly induced the expression of 35 proteins with 13 proteins showing an increasing trend. AMH downregulated only 3 proteins. ASH-induced downregulated proteins are involved in actin cytoskeleton maintenance needed for cell shape and migration which are critical for blood-brain barrier maintenance and angiogenesis. In contrast, ASH-induced upregulated proteins are RNA-binding proteins involved in RNA splicing, transport, and stability. Thus, ASH alters BMV proteomics to impair cytoskeletal integrity and RNA processing which are critical for cerebrovascular function.
Collapse
Affiliation(s)
- Siva SVP Sakamuri
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Venkata N Sure
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Lokanatha Oruganti
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - William Wisen
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
| | - Partha K Chandra
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
| | - Ning Liu
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, New Orleans, LA, USA
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Vivian A Fonseca
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Xiaoying Wang
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, New Orleans, LA, USA
- Department of Neurosurgery, Tulane University School of Medicine, New Orleans, LA, USA
| | - Jennifer Klein
- Department of Biochemistry & Molecular Biology, Louisiana State University School of Medicine, New Orleans, LA, USA
| | - Prasad VG Katakam
- Department of Pharmacology, Tulane University School of Medicine, New Orleans, LA, USA
- Neuroscience Program, Tulane Brain Institute, Tulane University, New Orleans, LA, USA
- Clinical Neuroscience Research Center, New Orleans, LA, USA
| |
Collapse
|
30
|
Wu Y, Sun J, Lin Q, Wang D, Hai J. Sustained release of vascular endothelial growth factor A and basic fibroblast growth factor from nanofiber membranes reduces oxygen/glucose deprivation-induced injury to neurovascular units. Neural Regen Res 2024; 19:887-894. [PMID: 37843225 PMCID: PMC10664103 DOI: 10.4103/1673-5374.382252] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 04/12/2023] [Accepted: 06/16/2023] [Indexed: 10/17/2023] Open
Abstract
Upregulation of vascular endothelial growth factor A/basic fibroblast growth factor (VEGFA/bFGF) expression in the penumbra of cerebral ischemia can increase vascular volume, reduce lesion volume, and enhance neural cell proliferation and differentiation, thereby exerting neuroprotective effects. However, the beneficial effects of endogenous VEGFA/bFGF are limited as their expression is only transiently increased. In this study, we generated multilayered nanofiber membranes loaded with VEGFA/bFGF using layer-by-layer self-assembly and electrospinning techniques. We found that a membrane containing 10 layers had an ideal ultrastructure and could efficiently and stably release growth factors for more than 1 month. This 10-layered nanofiber membrane promoted brain microvascular endothelial cell tube formation and proliferation, inhibited neuronal apoptosis, upregulated the expression of tight junction proteins, and improved the viability of various cellular components of neurovascular units under conditions of oxygen/glucose deprivation. Furthermore, this nanofiber membrane decreased the expression of Janus kinase-2/signal transducer and activator of transcription-3 (JAK2/STAT3), Bax/Bcl-2, and cleaved caspase-3. Therefore, this nanofiber membrane exhibits a neuroprotective effect on oxygen/glucose-deprived neurovascular units by inhibiting the JAK2/STAT3 pathway.
Collapse
Affiliation(s)
- Yifang Wu
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Sun
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qi Lin
- Department of Pharmacy, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dapeng Wang
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jian Hai
- Department of Neurosurgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| |
Collapse
|
31
|
Li X, Ding Y, Haddad YW, Geng X. Greater Omentum: Multifaceted Interactions in Neurological Recovery and Disease Progression. Aging Dis 2024; 15:2381-2394. [PMID: 38421824 PMCID: PMC11567243 DOI: 10.14336/ad.2024.0213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 02/13/2024] [Indexed: 03/02/2024] Open
Abstract
The greater omentum, a unique anatomical structure composed of adipocytes, loose connective tissue, and a dense vascular network. Plays a pivotal role beyond its traditional understanding. It houses specialized immunological units known as 'Milky spots,' making it a key player in immune response. Moreover, the omentum's capacity to enhance tissue perfusion, absorb edema fluid, boost acetylcholine synthesis, and foster neuron repair have rendered it a topic of interest in the context of various diseases, especially neurological disorders. This review provides a comprehensive overview of the intricate anatomy and histology of the greater omentum, casting light on its multifaceted functions and its associations with a spectrum of diseases. With a specific focus on neurological ailments, we delineate the intricate relationship that the omentum shares with other pathologies like stroke and we underly its contribution to serving as a therapeutic agent in neurological disorders. By deciphering the underlying mechanisms and emphasizing areas that demand further investigation. This review aims to spark renewed interest and pave the way for comprehensive studies exploring the greater omentum's potential in neurology and broader medicine overall. Given these diverse interactions that yet remain elusive, we must investigate and understand the nuanced relationship between the greater omentum and pathologies, especially its role in stroke's pathophysiology and therapeutic interventions so as to enhance patient care.
Collapse
Affiliation(s)
- Xiang Li
- Beijing Luhe Institute of Neuroscience, Capital Medical University, Beijing, China.
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Yazeed W. Haddad
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| | - Xiaokun Geng
- Beijing Luhe Institute of Neuroscience, Capital Medical University, Beijing, China.
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China.
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI 48201, USA.
| |
Collapse
|
32
|
Ren C, Li N, Xu J, Yang Y, Qin L, Jia L, Wang X, Huang S, Jin K, Liu F, Ji X, Li S. Hypoxic Conditioning: A Potential Perioperative Strategy to Reduce Abdominal Aortic Occlusion-Related Injury in Mouse Proximal and Distal Organs. Aging Dis 2024; 15:2863-2879. [PMID: 38300647 PMCID: PMC11567254 DOI: 10.14336/ad.2024.0121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 01/21/2024] [Indexed: 02/02/2024] Open
Abstract
This study aimed to investigate the impact of abdominal aortic occlusion (AAO)- induced injury on the kidney, lower limb muscles, heart, and brain in mice, and the potential protective effects of hypoxic postconditioning (HyC). The experimental design employed an abdominal aortic occlusion (AAO) model, and involved three groups of mice: sham, AAO, and AAO+HyC. Ten minutes after the AAO model, mice were subjected to hypoxic treatment lowering oxygen concentration to 5% within 45 minutes, and then returned to a normal oxygen environment. Hematoxylin- eosin (HE) stain was used for Histopathological examinations, and Quantibody Mouse Array was used for detecting apoptosis and inflammation-related protein expression. Histopathological examinations showed that HyC mitigated pathological damage to proximal organs (kidneys and lower limb muscles), distal organs (heart and brain), and reduced inflammatory cell infiltration. Expression of apoptosis- and inflammation-related proteins in brain and heart tissues were also evaluated. HyC significantly increased cellular inhibitor of apoptosis 2 (cIAP2) in the brain and Bcl-2 and insulin-like growth factor 2 (IGF-2) in the heart. Additionally, HyC regulated the expression of several inflammation-related factors in both brain and heart tissues. Although further investigation is needed, particularly in human subjects, this study highlights the potential of HyC as a promising therapeutic strategy for reducing AAO-associated organ damage.
Collapse
Affiliation(s)
- Changhong Ren
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Ning Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Jun Xu
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Yong Yang
- School of Chinese Medicine, Beijing University of Chines Medicine, Beijing, China
| | - Linhui Qin
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Linpei Jia
- Department of Nephrology, Xuanwu Hospital, Capital Medical University, Beijing, China.
| | - Xian Wang
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Shuangfeng Huang
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Kunlin Jin
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | - Fengyong Liu
- Department of Interventional Radiology, Senior Department of Oncology, Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Xunming Ji
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| | - Sijie Li
- Beijing Key Laboratory of Hypoxia Conditioning Translational Medicine, Xuanwu Hospital, Capital Medical University, Center of Stroke, Beijing Institute of Brain Disorder, Capital Medical University, Beijing, China.
| |
Collapse
|
33
|
Zhang M, Liu Q, Meng H, Duan H, Liu X, Wu J, Gao F, Wang S, Tan R, Yuan J. Ischemia-reperfusion injury: molecular mechanisms and therapeutic targets. Signal Transduct Target Ther 2024; 9:12. [PMID: 38185705 PMCID: PMC10772178 DOI: 10.1038/s41392-023-01688-x] [Citation(s) in RCA: 157] [Impact Index Per Article: 157.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Revised: 08/29/2023] [Accepted: 10/18/2023] [Indexed: 01/09/2024] Open
Abstract
Ischemia-reperfusion (I/R) injury paradoxically occurs during reperfusion following ischemia, exacerbating the initial tissue damage. The limited understanding of the intricate mechanisms underlying I/R injury hinders the development of effective therapeutic interventions. The Wnt signaling pathway exhibits extensive crosstalk with various other pathways, forming a network system of signaling pathways involved in I/R injury. This review article elucidates the underlying mechanisms involved in Wnt signaling, as well as the complex interplay between Wnt and other pathways, including Notch, phosphatidylinositol 3-kinase/protein kinase B, transforming growth factor-β, nuclear factor kappa, bone morphogenetic protein, N-methyl-D-aspartic acid receptor-Ca2+-Activin A, Hippo-Yes-associated protein, toll-like receptor 4/toll-interleukine-1 receptor domain-containing adapter-inducing interferon-β, and hepatocyte growth factor/mesenchymal-epithelial transition factor. In particular, we delve into their respective contributions to key pathological processes, including apoptosis, the inflammatory response, oxidative stress, extracellular matrix remodeling, angiogenesis, cell hypertrophy, fibrosis, ferroptosis, neurogenesis, and blood-brain barrier damage during I/R injury. Our comprehensive analysis of the mechanisms involved in Wnt signaling during I/R reveals that activation of the canonical Wnt pathway promotes organ recovery, while activation of the non-canonical Wnt pathways exacerbates injury. Moreover, we explore novel therapeutic approaches based on these mechanistic findings, incorporating evidence from animal experiments, current standards, and clinical trials. The objective of this review is to provide deeper insights into the roles of Wnt and its crosstalk signaling pathways in I/R-mediated processes and organ dysfunction, to facilitate the development of innovative therapeutic agents for I/R injury.
Collapse
Affiliation(s)
- Meng Zhang
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
| | - Qian Liu
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hui Meng
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Hongxia Duan
- Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Xin Liu
- Second Clinical Medical College, Jining Medical University, Jining, Shandong, 272067, China
| | - Jian Wu
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Fei Gao
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China
- Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Shijun Wang
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital and Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| | - Rubin Tan
- Department of Physiology, Basic medical school, Xuzhou Medical University, Xuzhou, 221004, China.
| | - Jinxiang Yuan
- The Collaborative Innovation Center, Jining Medical University, Jining, Shandong, 272067, China.
| |
Collapse
|
34
|
Wang G, Li Z, Lin P, Zhang H, Wang Y, Zhang T, Wang H, Li H, Lin L, Zhao Y, Jia L, Chen Y, Ji H, Zhao W, Fu Z, Zhong Z. Knockdown of Smox protects the integrity of the blood-brain barrier through antioxidant effect and Nrf2 pathway activation in stroke. Int Immunopharmacol 2024; 126:111183. [PMID: 37984250 DOI: 10.1016/j.intimp.2023.111183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 10/23/2023] [Accepted: 11/01/2023] [Indexed: 11/22/2023]
Abstract
Once an ischemic stroke occurs, reactive oxygen species (ROS) and oxidative stress degrade the tight connections between cerebral endothelial cells resulting in their damage. The expression of antioxidant genes may be enhanced, and ROS formation may be reduced following Nrf2 activation, which is associated with protection against ischemic stroke. Overexpression of spermine oxidase (Smox) in the neocortex led to increased H2O2 production. However, how Smox impacts the regulation of the blood-brain barrier (BBB) through antioxidants has not been examined yet. We conducted experiments both in the cell level and in the transient middle cerebral artery occlusion (tMCAO) model to evaluate the effect of Smox siRNA lentivirus (si-Smox) knockdown on BBB protection against ischemic stroke. Mice treated with si-Smox showed remarkably decreased BBB breakdown and reduced endothelial inflammation following stroke. The treatment with si-Smox significantly elevated the Bcl-2 to Bax ratio and decreased the production of cleaved caspase-3 in the tMCAO model. Further investigation revealed that the neuroprotective effect was the result of the antioxidant properties of si-Smox, which reduced oxidative stress and enhanced CD31+ cells in the peri-infarct cortical areas. Of significance, si-Smox activated Nrf2 in both bEnd.3 cells and tMCAO animals, and blocking Nrf2 with brusatol diminished the protective effects of si-Smox. The study findings suggest that si-Smox exerts neuroprotective effects and promotes angiogenesis by activating the Nrf2 pathway, thus decreasing oxidative stress and apoptosis caused by tMCAO. As a result, si-Smox may hold potential as a therapeutic candidate for preserving BBB integrity while treating ischemic stroke.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China
| | - Peng Lin
- Department of Biochemistry and Molecular Biology, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hui Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Heming Li
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Lexun Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yuehui Zhao
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Lina Jia
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Yang Chen
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Hong Ji
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Wenran Zhao
- Department of Cell Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China
| | - Zhongqiu Fu
- Department of Neonatology, Zhuhai Women and Children's Hospital, Zhuhai, Guangdong 519000, China.
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang 150081, China.
| |
Collapse
|
35
|
Wang Q, Kohls W, Wills M, Li F, Pang Q, Geng X, Ding Y. A novel stroke rehabilitation strategy and underlying stress granule regulations through inhibition of NLRP3 inflammasome activation. CNS Neurosci Ther 2024; 30:e14405. [PMID: 37580991 PMCID: PMC10805392 DOI: 10.1111/cns.14405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/05/2023] [Accepted: 07/23/2023] [Indexed: 08/16/2023] Open
Abstract
OBJECTIVE Dynamic changes in ischemic pathology after stroke suggested a "critical window" of enhanced neuroplasticity immediately after stroke onset. Although physical exercise has long been considered a promising strategy of stroke rehabilitation, very early physical exercise may exacerbate brain injury. Since remote ischemic conditioning (RIC) promotes neuroprotection and neuroplasticity, the present study combined RIC with sequential exercise to establish a new rehabilitation strategy for a better rehabilitative outcome. METHODS A total of 120 adult male Sprague-Dawley rats were used and divided into five groups: (1) sham, (2) stroke, (3) stroke with exercise, (4) stroke with RIC, and (5) stroke with RIC followed by exercise. Brain damage was evaluated by infarct volume, neurological deficit, cell death, and lactate dehydrogenase (LDH) activity. Long-term functional outcomes were determined by grid walk tests, rotarod tests, beam balance tests, forelimb placing tests, and the Morris water maze. Neuroplasticity was evaluated through measurements of both mRNA and protein levels of synaptogenesis (synaptophysin [SYN], post-synaptic density protein-95 [PSD-95], and brain-derived neurotrophic factor [BDNF]) and angiogenesis (vascular endothelial growth factor [VEGF], angiopoietin-1 [Ang-1], and angiopoietin-2 [Ang-2]). Inflammasome activation was measured by concentrations of interleukin-18 (IL-18) and IL-1β detected by enzyme-linked immunosorbent assay (ELISA) kits, mRNA expressions of NLR pyrin domain containing 3 (NLRP3), apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain (ASC), IL-18 and IL-1β, and protein quantities of NLRP3, ASC, cleaved-caspase-1, gasdermin D-N (GSDMD-N), and IL-18 and IL-1β. Stress granules (SGs), including GTPase-activating protein-binding protein 1 (G3BP1), T cell-restricted intracellular antigen-1 (TIA1), and DEAD-box RNA helicase 3X (DDX3X) were evaluated at mRNA and protein levels. The interactions between DDX3X with NLRP3 or G3BP1 were determined by immunofluorescence and co-immunoprecipitation. RESULTS Early RIC decreased infarct volumes, neurological deficits, cell death, and LDH activity at post-stroke Day 3 (p < 0.05). All treatment groups showed significant improvement in functional outcomes, including sensory, motor, and cognitive functions. RIC and exercise, as compared to RIC or physical exercise alone, had improved functional outcomes after stroke (p < 0.05), as well as synaptogenesis and angiogenesis (p < 0.05). RIC significantly reduced mRNA and protein expressions of NLRP3 (p < 0.05). SGs formation peaked at 0 h after ischemia, then progressively decreased until 24 h postreperfusion, which was reversed by RIC (p < 0.05). The assembly of SGs consumed DDX3X and then inhibited NLRP3 inflammasome activation. CONCLUSIONS RIC followed by exercise induced a better rehabilitation in ischemic rats, while early RIC alleviated ischemia-reperfusion injury via stress-granule-mediated inhibition of NLRP3 inflammasome.
Collapse
Affiliation(s)
- Qingzhu Wang
- China‐America Institute of NeuroscienceBeijing Luhe Hospital, Capital Medical UniversityBeijingChina
| | - Wesley Kohls
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| | - Melissa Wills
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| | - Fengwu Li
- China‐America Institute of NeuroscienceBeijing Luhe Hospital, Capital Medical UniversityBeijingChina
| | - Qi Pang
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
- Department of Neurosurgery, Shandong Provincial HospitalShandong UniversityJinanChina
| | - Xiaokun Geng
- China‐America Institute of NeuroscienceBeijing Luhe Hospital, Capital Medical UniversityBeijingChina
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
- Department of Neurology, Beijing Luhe HospitalCapital Medical UniversityBeijingChina
| | - Yuchuan Ding
- Department of NeurosurgeryWayne State University School of MedicineDetroitMichiganUSA
| |
Collapse
|
36
|
Qi L, Wang F, Sun X, Li H, Zhang K, Li J. Recent advances in tissue repair of the blood-brain barrier after stroke. J Tissue Eng 2024; 15:20417314241226551. [PMID: 38304736 PMCID: PMC10832427 DOI: 10.1177/20417314241226551] [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: 07/26/2023] [Accepted: 12/31/2023] [Indexed: 02/03/2024] Open
Abstract
The selective permeability of the blood-brain barrier (BBB) enables the necessary exchange of substances between the brain parenchyma and circulating blood and is important for the normal functioning of the central nervous system. Ischemic stroke inflicts damage upon the BBB, triggering adverse stroke outcomes such as cerebral edema, hemorrhagic transformation, and aggravated neuroinflammation. Therefore, effective repair of the damaged BBB after stroke and neovascularization that allows for the unique selective transfer of substances from the BBB after stroke is necessary and important for the recovery of brain function. This review focuses on four important therapies that have effects of BBB tissue repair after stroke in the last seven years. Most of these new therapies show increased expression of BBB tight-junction proteins, and some show beneficial results in terms of enhanced pericyte coverage at the injured vessels. This review also briefly outlines three effective classes of approaches and their mechanisms for promoting neoangiogenesis following a stroke.
Collapse
Affiliation(s)
- Liujie Qi
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Fei Wang
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Xiaojing Sun
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Hang Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| | - Kun Zhang
- School of Life Science, Zhengzhou University, Zhengzhou, PR China
| | - Jingan Li
- School of Material Science and Engineering & Henan Key Laboratory of Advanced Magnesium Alloy & Key Laboratory of Materials Processing and Mold (Ministry of Education), Zhengzhou University, Zhengzhou, PR China
| |
Collapse
|
37
|
Tian T, Wang L, Xu J, Jia Y, Xue K, Huang S, Shen T, Luo Y, Li S, Min L. Prediction of early neurological deterioration in acute ischemic stroke patients treated with intravenous thrombolysis. J Cereb Blood Flow Metab 2023; 43:2049-2059. [PMID: 37668997 PMCID: PMC10925869 DOI: 10.1177/0271678x231200117] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/06/2023] [Accepted: 08/13/2023] [Indexed: 09/06/2023]
Abstract
A proportion of acute ischemic stroke (AIS) patients suffer from early neurological deterioration (END) within 24 hours following intravenous thrombolysis (IVT), which greatly increases the risk of poor prognosis of these patients. Therefore, we aimed to explore the predictors of early neurological deterioration of ischemic origin (ENDi) in AIS patients after IVT and develop a nomogram prediction model. This study collected 244 AIS patients with post-thrombolysis ENDi as the derivation cohort and 155 patients as the validation cohort. To establish a nomogram prediction model, risk factors were identified by multivariate logistic regression analysis. The results showed that neutrophil to lymphocyte ratio (NLR) (OR 2.616, 95% CI 1.640-4.175, P < 0.001), mean platelet volume (MPV) (OR 3.334, 95% CI 1.351-8.299, P = 0.009), body mass index (BMI) (OR 1.979, 95% CI 1.285-3.048, P = 0.002) and atrial fibrillation (AF) (OR 8.012, 95% CI 1.341-47.873, P = 0.023) were significantly associated with ENDi. The area under the curve of the prediction model constructed from the above four factors was 0.981 (95% CI 0.961-1.000) and the calibration curve was close to the ideal diagonal line. Therefore, this nomogram prediction model exhibited good discrimination and calibration power and might be a reliable and easy-to-use tool to predict post-thrombolysis ENDi in AIS patients.
Collapse
Affiliation(s)
- Tian Tian
- Department of Neurology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Lanjing Wang
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jiali Xu
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yujie Jia
- Department of Neurology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Kun Xue
- Department of Neurology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| | - Shuangfeng Huang
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tong Shen
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yumin Luo
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Sijie Li
- Department of Emergency, Xuanwu Hospital, Capital Medical University, Beijing, China
- Department of Neurology, Xuanwu Hospital, Capital Medical University, Beijing, China
- Beijing Institute of Brain Disorders, Collaborative Innovation Center for Brain Disorders, Capital Medical University, Beijing, China
| | - Lianqiu Min
- Department of Neurology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou, China
| |
Collapse
|
38
|
Jiang X, Zhou L, Sun Z, Xie B, Lin H, Gao X, Deng L, Yang C. MSCs overexpressing GDNF restores brain structure and neurological function in rats with intracerebral hemorrhage. MOLECULAR BIOMEDICINE 2023; 4:43. [PMID: 38008847 PMCID: PMC10678901 DOI: 10.1186/s43556-023-00159-7] [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: 08/06/2023] [Accepted: 11/16/2023] [Indexed: 11/28/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have been applied in transplantation to treat intracerebral hemorrhage (ICH) but with limited efficacy. Accumulated evidence has shown that glial cell-derived neurotrophic factor (GDNF) plays a crucial part in neuronal protection and functional recovery of the brain after ICH; however, GDNF has difficulty crossing the blood-brain barrier, which limits its application. In this study, we investigated the influences of MSCs overexpressing GDNF (MSCs/GDNF) on the brain structure as well as gait of rats after ICH and explored the possible mechanisms. We found that cell transplantation could reverse the neurological dysfunction and brain damage caused by ICH to a certain extent, and MSCs/GDNF transplantation was superior to MSCs transplantation. Moreover, Transplantation of MSCs overexpressing GDNF effectively reduced the volume of bleeding foci and increased the level of glucose uptake in rats with ICH, which could be related to improving mitochondrial quality. Furthermore, GDNF produced by transplanted MSCs/GDNF further inhibited neuroinflammation, improved mitochondrial quality and function, promoted angiogenesis and the survival of neurons and oligodendrocytes, and enhanced synaptic plasticity in ICH rats when compared with simple MSC transplantation. Overall, our data indicate that GDNF overexpression heightens the curative effect of MSC implantation in treating rats following ICH.
Collapse
Affiliation(s)
- Xiaoqian Jiang
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ling Zhou
- Clinical Skills Center, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zihuan Sun
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Bingqing Xie
- Laboratory of Neurological Diseases and Brain Function, the Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
- Institute of Epigenetics and Brain Science, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Heng Lin
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Xiaoqing Gao
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Li Deng
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Chaoxian Yang
- Department of Anatomy, College of Basic Medicine, Southwest Medical University, Luzhou, 646000, Sichuan, China.
- Department of Neurobiology, Preclinical Medicine Research Center, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
39
|
Lin W, Zhao XY, Cheng JW, Li LT, Jiang Q, Zhang YX, Han F. Signaling pathways in brain ischemia: Mechanisms and therapeutic implications. Pharmacol Ther 2023; 251:108541. [PMID: 37783348 DOI: 10.1016/j.pharmthera.2023.108541] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 09/18/2023] [Accepted: 09/25/2023] [Indexed: 10/04/2023]
Abstract
Ischemic stroke occurs when the arteries supplying blood to the brain are narrowed or blocked, inducing damage to brain tissue due to a lack of blood supply. One effective way to reduce brain damage and alleviate symptoms is to reopen blocked blood vessels in a timely manner and reduce neuronal damage. To achieve this, researchers have focused on identifying key cellular signaling pathways that can be targeted with drugs. These pathways include oxidative/nitrosative stress, excitatory amino acids and their receptors, inflammatory signaling molecules, metabolic pathways, ion channels, and other molecular events involved in stroke pathology. However, evidence suggests that solely focusing on protecting neurons may not yield satisfactory clinical results. Instead, researchers should consider the multifactorial and complex mechanisms underlying stroke pathology, including the interactions between different components of the neurovascular unit. Such an approach is more representative of the actual pathological process observed in clinical settings. This review summarizes recent research on the multiple molecular mechanisms and drug targets in ischemic stroke, as well as recent advances in novel therapeutic strategies. Finally, we discuss the challenges and future prospects of new strategies based on the biological characteristics of stroke.
Collapse
Affiliation(s)
- Wen Lin
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Xiang-Yu Zhao
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Jia-Wen Cheng
- Department of Physiology, School of Basic Medical Sciences, Nanjing Medical University, Nanjing 211166, China
| | - Li-Tao Li
- Department of Neurology, Hebei General Hospital, Shijiazhuang 050051, Hebei, China
| | - Quan Jiang
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 14853, USA
| | - Yi-Xuan Zhang
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China.
| | - Feng Han
- Medical Basic Research Innovation Center for Cardiovascular and Cerebrovascular Diseases, Ministry of Education, China; International Joint Laboratory for Drug Target of Critical Illnesses, Key Laboratory of Cardiovascular and Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China; Gusu School, Nanjing Medical University, Suzhou Municipal Hospital, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou 215002, China; Institute of Brain Science, the Affiliated Brain Hospital of Nanjing Medical University, Nanjing 211166, China.
| |
Collapse
|
40
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
41
|
Huang Y, Omorou M, Gao M, Mu C, Xu W, Xu H. Hydrogen sulfide and its donors for the treatment of cerebral ischaemia-reperfusion injury: A comprehensive review. Biomed Pharmacother 2023; 161:114506. [PMID: 36906977 DOI: 10.1016/j.biopha.2023.114506] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
As an endogenous gas signalling molecule, hydrogen sulfide (H2S) is frequently present in a variety of mammals and plays a significant role in the cardiovascular and nervous systems. Reactive oxygen species (ROS) are produced in large quantities as a result of cerebral ischaemia-reperfusion, which is a very serious class of cerebrovascular diseases. ROS cause oxidative stress and induce specific gene expression that results in apoptosis. H2S reduces cerebral ischaemia-reperfusion-induced secondary injury via anti-oxidative stress injury, suppression of the inflammatory response, inhibition of apoptosis, attenuation of cerebrovascular endothelial cell injury, modulation of autophagy, and antagonism of P2X7 receptors, and it plays an important biological role in other cerebral ischaemic injury events. Despite the many limitations of the hydrogen sulfide therapy delivery strategy and the difficulty in controlling the ideal concentration, relevant experimental evidence demonstrating that H2S plays an excellent neuroprotective role in cerebral ischaemia-reperfusion injury (CIRI). This paper examines the synthesis and metabolism of the gas molecule H2S in the brain as well as the molecular mechanisms of H2S donors in cerebral ischaemia-reperfusion injury and possibly other unknown biological functions. With the active development in this field, it is expected that this review will assist researchers in their search for the potential value of hydrogen sulfide and provide new ideas for preclinical trials of exogenous H2S.
Collapse
Affiliation(s)
- Yiwei Huang
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| | - Moussa Omorou
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Meng Gao
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Chenxi Mu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Weijing Xu
- School of Public Health, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Hui Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| |
Collapse
|
42
|
Lavayen BP, Yang C, Larochelle J, Liu L, Tishko RJ, de Oliveira ACP, Muñoz E, Candelario-Jalil E. Neuroprotection by the cannabidiol aminoquinone VCE-004.8 in experimental ischemic stroke in mice. Neurochem Int 2023; 165:105508. [PMID: 36863495 DOI: 10.1016/j.neuint.2023.105508] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/20/2023] [Accepted: 02/27/2023] [Indexed: 03/04/2023]
Abstract
Synthetic cannabidiol (CBD) derivative VCE-004.8 is a peroxisome proliferator-activated receptor gamma (PPARγ) and cannabinoid receptor type 2 (CB2) dual agonist with hypoxia mimetic activity. The oral formulation of VCE-004.8, termed EHP-101, possesses anti-inflammatory properties and is currently in phase 2 clinical trials for relapsing forms of multiple sclerosis. The activation of PPARγ or CB2 receptors exerts neuroprotective effects by dampening neuroinflammation in ischemic stroke models. However, the effect of a dual PPARγ/CB2 agonist in ischemic stroke models is not known. Here, we demonstrate that treatment with VCE-004.8 confers neuroprotection in young mice subjected to cerebral ischemia. Male C57BL/6J mice, aged 3-4 months, were subjected to 30-min transient middle cerebral artery occlusion (MCAO). We evaluated the effect of intraperitoneal VCE-004.8 treatment (10 or 20 mg/kg) either at the onset of reperfusion or 4h or 6h after the reperfusion. Seventy-two hours after ischemia, animals were subjected to behavioral tests. Immediately after the tests, animals were perfused, and brains were collected for histology and PCR analysis. Treatment with VCE-004.8 either at the onset or 4h after reperfusion significantly reduced infarct volume and improved behavioral outcomes. A trend toward reduction in stroke injury was observed in animals receiving the drug starting 6h after recirculation. VCE-004.8 significantly reduced the expression of pro-inflammatory cytokines and chemokines involved in BBB breakdown. Mice receiving VCE-004.8 had significantly lower levels of extravasated IgG in the brain parenchyma, indicating protection against stroke-induced BBB disruption. Lower levels of active matrix metalloproteinase-9 were found in the brain of drug-treated animals. Our data show that VCE-004.8 is a promising drug candidate for treating ischemic brain injury. Since VCE-004.8 has been shown to be safe in the clinical setting, the possibility of repurposing its use as a delayed treatment option for ischemic stroke adds substantial translational value to our findings.
Collapse
Affiliation(s)
- Bianca P Lavayen
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Changjun Yang
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Jonathan Larochelle
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Lei Liu
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Ryland J Tishko
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA
| | - Antonio Carlos Pinheiro de Oliveira
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA; Neuropharmacology Laboratory, Department of Pharmacology, Universidade Federal de Minas Gerais, Brazil
| | - Eduardo Muñoz
- Instituto Maimónides de Investigación Biomédica de Córdoba-IMIBIC, Córdoba, Spain; Departamento de Biología Celular, Fisiología e Inmunología, Universidad de Córdoba, Córdoba, Spain; Hospital Universitario Reina Sofía, Córdoba, Spain
| | - Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
43
|
Bui TA, Jickling GC, Winship IR. Neutrophil dynamics and inflammaging in acute ischemic stroke: A transcriptomic review. Front Aging Neurosci 2022; 14:1041333. [PMID: 36620775 PMCID: PMC9813499 DOI: 10.3389/fnagi.2022.1041333] [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: 09/10/2022] [Accepted: 11/28/2022] [Indexed: 12/24/2022] Open
Abstract
Stroke is among the leading causes of death and disability worldwide. Restoring blood flow through recanalization is currently the only acute treatment for cerebral ischemia. Unfortunately, many patients that achieve a complete recanalization fail to regain functional independence. Recent studies indicate that activation of peripheral immune cells, particularly neutrophils, may contribute to microcirculatory failure and futile recanalization. Stroke primarily affects the elderly population, and mortality after endovascular therapies is associated with advanced age. Previous analyses of differential gene expression across injury status and age identify ischemic stroke as a complex age-related disease. It also suggests robust interactions between stroke injury, aging, and inflammation on a cellular and molecular level. Understanding such interactions is crucial in developing effective protective treatments. The global stroke burden will continue to increase with a rapidly aging human population. Unfortunately, the mechanisms of age-dependent vulnerability are poorly defined. In this review, we will discuss how neutrophil-specific gene expression patterns may contribute to poor treatment responses in stroke patients. We will also discuss age-related transcriptional changes that may contribute to poor clinical outcomes and greater susceptibility to cerebrovascular diseases.
Collapse
Affiliation(s)
- Truong An Bui
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Glen C. Jickling
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Medicine, Division of Neurology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Ian R. Winship
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
44
|
Zhang Y, Zhao X, Zhang Y, Zeng F, Yan S, Chen Y, Li Z, Zhou D, Liu L. The role of circadian clock in astrocytes: From cellular functions to ischemic stroke therapeutic targets. Front Neurosci 2022; 16:1013027. [PMID: 36570843 PMCID: PMC9772621 DOI: 10.3389/fnins.2022.1013027] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 11/10/2022] [Indexed: 12/14/2022] Open
Abstract
Accumulating evidence suggests that astrocytes, the abundant cell type in the central nervous system (CNS), play a critical role in maintaining the immune response after cerebral infarction, regulating the blood-brain barrier (BBB), providing nutrients to the neurons, and reuptake of glutamate. The circadian clock is an endogenous timing system that controls and optimizes biological processes. The central circadian clock and the peripheral clock are consistent, controlled by various circadian components, and participate in the pathophysiological process of astrocytes. Existing evidence shows that circadian rhythm controls the regulation of inflammatory responses by astrocytes in ischemic stroke (IS), regulates the repair of the BBB, and plays an essential role in a series of pathological processes such as neurotoxicity and neuroprotection. In this review, we highlight the importance of astrocytes in IS and discuss the potential role of the circadian clock in influencing astrocyte pathophysiology. A comprehensive understanding of the ability of the circadian clock to regulate astrocytes after stroke will improve our ability to predict the targets and biological functions of the circadian clock and gain insight into the basis of its intervention mechanism.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Xin Zhao
- The Medical School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhang
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Fukang Zeng
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,The Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Siyang Yan
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yao Chen
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Zhong Li
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Desheng Zhou
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,Desheng Zhou,
| | - Lijuan Liu
- Department of Neurology, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China,*Correspondence: Lijuan Liu,
| |
Collapse
|
45
|
Neuroprotective Effect of Polyphenol Extracts from Terminalia chebula Retz. against Cerebral Ischemia-Reperfusion Injury. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27196449. [PMID: 36234986 PMCID: PMC9571999 DOI: 10.3390/molecules27196449] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/22/2022] [Accepted: 09/26/2022] [Indexed: 11/17/2022]
Abstract
Current therapies for ischemic stroke are insufficient due to the lack of specific drugs. This study aimed to investigate the protective activity of polyphenol extracts from Terminalia chebula against cerebral ischemia-reperfusion induced damage. Polyphenols of ethyl acetate and n-butanol fractions were extracted from T. chebula. BV2 microglial cells exposed to oxygen-glucose deprivation/reoxygenation and mice subjected to middle cerebral artery occlusion/reperfusion were treated by TPE and TPB. Cell viability, cell morphology, apoptosis, mitochondrial membrane potential, enzyme activity and signaling pathway related to oxidative stress were observed. We found that TPE and TPB showed strong antioxidant activity in vitro. The protective effects of TPE and TPB on cerebral ischemia-reperfusion injury were demonstrated by enhanced antioxidant enzyme activities, elevated level of the nucleus transportation of nuclear factor erythroid 2-related factor 2 and expressions of antioxidant proteins, with a simultaneous reduction in cell apoptosis and reactive oxygen species level. In conclusion, TPE and TPB exert neuroprotective effects by stimulating the Nrf2 signaling pathway, thereby inhibiting apoptosis.
Collapse
|
46
|
Nrf2 Pathway and Autophagy Crosstalk: New Insights into Therapeutic Strategies for Ischemic Cerebral Vascular Diseases. Antioxidants (Basel) 2022; 11:antiox11091747. [PMID: 36139821 PMCID: PMC9495910 DOI: 10.3390/antiox11091747] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/27/2022] [Accepted: 08/30/2022] [Indexed: 11/17/2022] Open
Abstract
Cerebrovascular disease is highly prevalent and has a complex etiology and variable pathophysiological activities. It thus poses a serious threat to human life and health. Currently, pathophysiological research on cerebrovascular diseases is gradually improving, and oxidative stress and autophagy have been identified as important pathophysiological activities that are gradually attracting increasing attention. Many studies have found some effects of oxidative stress and autophagy on cerebrovascular diseases, and studies on the crosstalk between the two in cerebrovascular diseases have made modest progress. However, further, more detailed studies are needed to determine the specific mechanisms. This review discusses nuclear factor erythroid 2-related factor 2 (Nrf2) molecules, which are closely associated with oxidative stress and autophagy, and the crosstalk between them, with the aim of providing clues for studying the two important pathophysiological changes and their crosstalk in cerebrovascular diseases as well as exploring new target treatments.
Collapse
|
47
|
Ibrahim A, Badr A, Amer HA, Eidaroos AS, Mariol N, Mesalam A. Vascular endothelial growth factor: A novel marker for torsion-induced incomplete cervical dilatation in Egyptian buffaloes. Open Vet J 2022; 12:709-717. [PMID: 36589390 PMCID: PMC9789758 DOI: 10.5455/ovj.2022.v12.i5.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 08/22/2022] [Indexed: 11/06/2022] Open
Abstract
Background Uterine torsion, the most complicated cause of maternal dystocia recorded in bovine, usually followed by incomplete cervical dilatation after successful detorsion, which interfere with vaginal fetal delivery. Aim This study aimed to evaluate the histopathological changes, variable collagen concentrations, and vascular endothelial growth factor expressions in uterine and cervical tissues following successful detorsion. Methods Animals were classified into four groups; normally calved cases (group A), cases that respond successfully to detorsion and followed by vaginal fetal delivery without (group B) or with (group C) cervical laceration, and cases that suffered from failure of complete cervical dilatation after successful detorsion (group D). Results Histopathological findings revealed variable changes in all uterine torsion affected groups, which were characteristic and marked in animals that suffered from failure of complete cervical dilatation following successful detorsion. Moreover, failure of cervical dilatation was associated with the highest collagen concentrations as shown by Masson trichrome stain. On the other hand, immunohistochemical findings showed that the normally calved cases have the highest vascular endothelial growth factor expression compared with animals that suffered from failure of complete cervical dilatation. Conclusion Our results showed that the vascular endothelial growth factor is essential for cervical dilatation and its lower expression is accompanied by incomplete cervical dilatation following successful detorsion.
Collapse
Affiliation(s)
- Ahmed Ibrahim
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Atef Badr
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Hussein Ahmed Amer
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Abdel-Salam Eidaroos
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Najmi Mariol
- Department of Surgery and Theriogenology, Faculty of Veterinary Medicine, University of Tripoli, Tripoli, Libya
| | - Ayman Mesalam
- Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt,Corresponding Author: Ayman Mesalam. Department of Theriogenology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt.
| |
Collapse
|