1
|
Sun P, Xu Y, Xiong T, Li S, Qiu N, Zhou C, Wang J, Chang A, Chandran UR, Yin KJ. Genetic deletion of microRNA-15a/16-1 in pericytes stimulates cerebral angiogenesis and promotes functional recovery after ischemic stroke. Angiogenesis 2025; 28:35. [PMID: 40526299 DOI: 10.1007/s10456-025-09987-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 06/09/2025] [Indexed: 06/19/2025]
Abstract
Stroke is a leading cause of mortality and disability globally. Despite advancements in acute stroke therapies, patient outcomes with ischemic stroke remain suboptimal. Understanding its molecular mechanisms is crucial for developing effective treatments. Angiogenesis actively contributes to post-stroke functional recovery and improves long-term survival in stroke patients. Pericytes are essential for maintaining vascular stability and promoting angiogenesis. We hypothesized that microRNA-15a/16-1 in pericytes significantly modulates post-stroke angiogenesis and neurological recovery. Using a pericyte-specific miR-15a/16-1 conditional knockout (cKO) mouse model, we found that genetic deletion of miR-15a/16-1 in pericytes enhances angiogenesis, promotes cerebral blood flow recovery, and improves sensorimotor and cognitive outcomes following ischemic stroke. Mechanistically, RNA sequencing identified several novel targets of miR-15a/16-1, including Pappa2, Fgf9, Islr, and Ccr2. Interestingly, Pappa2, Fgf9, and Islr function as secreted proteins. Luciferase reporter assays demonstrated that miR-15a/16-1 directly binds and suppresses Pappa2, Fgf9, Islr, and Ccr2 activity in cultured pericytes. In vivo and in vitro assays further confirmed that miR-15a/16-1 silencing in pericytes significantly elevates the protein levels of Pappa2, Fgf9, Islr, and Ccr2 and enhances endothelial cell proliferation, migration, and tube formation under ischemic conditions. These findings suggest that targeting miR-15a/16-1 in pericytes offers a promising therapeutic strategy for enhancing stroke recovery by promoting neurovascular repair and reducing brain damage.
Collapse
Affiliation(s)
- Ping Sun
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Yang Xu
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tianqing Xiong
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shun Li
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Na Qiu
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| | - Chao Zhou
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jiefei Wang
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexander Chang
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Uma R Chandran
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ke-Jie Yin
- Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA.
| |
Collapse
|
2
|
Wang B, Li D, Peng C, Hong J, Wu Y. Dietary omega-3 intake and cognitive function in older adults. Int J Psychiatry Med 2025; 60:265-279. [PMID: 39277856 DOI: 10.1177/00912174241284925] [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] [Indexed: 09/17/2024]
Abstract
ObjectiveDietary habits have long been known to be a critical factor influencing cognitive health, especially among older adults. Despite extensive research on various dietary supplements, the impact of omega-3 polyunsaturated fatty acids (PUFAs) on cognitive function has not yet been thoroughly investigated. This research seeks to determine whether taking more omega-3 PUFAs correlates with improved cognitive function in older adults.MethodsCross-sectional data were analyzed from the National Health and Nutrition Examination Survey (NHANES), which included 2430 elderly participants aged 60 and above in the United States. The association between omega-3 consumption and cognitive outcomes was evaluated using linear regression models. Smoothing curves and threshold effect analysis were employed to examine nonlinear associations. Subgroup studies were conducted to demonstrate the strength and reliability of the association and factors affecting it.ResultsThe fully adjusted model demonstrated significant positive correlations between omega-3 intake and scores on all 3 cognitive assessments. Specifically, in the final model, the beta coefficients for the CERAD Word Learning test, Animal Fluency Test, and Digit Symbol Substitution Test were 0.53 (95% CI: 0.33-0.72, P < 0.0001), 0.29 (95% CI: 0.12-0.47, P = 0.001), and 0.61 (95% CI: 0.19-1.03, P = 0.0045), respectively.ConclusionIncreased intake of omega-3 was positively and independently associated with cognitive function in older adults, suggesting that consumption of omega-3 PUFAs may help to prevent cognitive decline with aging. Prospective studies are needed to determine the direct of effect in this association.
Collapse
Affiliation(s)
- Bingdian Wang
- School of Nursing, Anhui Medical University, Hefei, China
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Deqin Li
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Cuicui Peng
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jingfang Hong
- School of Nursing, Anhui Medical University, Hefei, China
| | - Yonggui Wu
- School of Nursing, Anhui Medical University, Hefei, China
- Department of Nephropathy, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Andone S, Lénárd F, Imre S, Dumitreasa M, Bălașa R. Fatty Acid Ratios Versus Conventional Risk Factors in Stroke: Insights into Severe Disability and Mortality Outcomes. Nutrients 2025; 17:1518. [PMID: 40362827 PMCID: PMC12073307 DOI: 10.3390/nu17091518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2025] [Revised: 04/20/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Objective: This study aims to investigate the role of fatty acid ratios, specifically DHA/ARA and EPA/ARA, in predicting severe disability and mortality in stroke patients and compare these ratios with conventional risk factors such as age, sex, hypertension, diabetes, and dyslipidemia. Methods: A prospective study was conducted involving 298 consecutive acute ischemic stroke patients (within 72 h of onset). Fatty acid ratios were measured from plasma, and all patients' evolution was followed through hospitalization. Binary logistic regression analysis was used to identify predictors of severe disability at discharge (Rankin 4-6) and in-hospital mortality, including fatty acid ratios and conventional risk factors. Results: A higher DHA/ARA ratio was associated with a reduced chance of severe disability (OR = 0.81), while a higher EPA/ARA ratio was associated with an increased chance of severe disability (OR = 1.70). Age was a significant factor, with older age (median 70 years) associated with a lower survivability chance (OR = 0.93) and a higher likelihood of severe disability when surviving. Fatty acid ratios did not significantly affect mortality outcomes. For male patients, EPA/AA ratios showed a powerful association with severe disability (p = 0.045), while no significant effect of fatty acids was observed in females. Conclusions: Fatty acids were significant predictors of severe disability in patients with acute ischemic stroke, independent of conventional risk factors, but without having any effect on in-hospital mortality. Age remained the only significant conventional risk factor predictor of outcome. Integrating fatty acid ratios alongside conventional risk factors may improve predictions of severe post-stroke disability, potentially guiding more personalized interventions for stroke patients.
Collapse
Affiliation(s)
- Sebastian Andone
- Ist Neurology Clinic, Emergency Clinical County Hospital Târgu Mureș, 540136 Târgu Mureș, Romania
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania
| | - Farczádi Lénárd
- Center for Advanced Medical and Pharmaceutical Research, ‘George Emil Palade’ University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania
| | - Silvia Imre
- Center for Advanced Medical and Pharmaceutical Research, ‘George Emil Palade’ University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania
- Department of Analytical Chemistry and Drug Analysis, Faculty of Pharmacy, ‘George Emil Palade’ University of Medicine, Pharmacy, Science, and Technology of Târgu Mures, 540136 Târgu Mures, Romania
| | - Mihai Dumitreasa
- Ist Neurology Clinic, Emergency Clinical County Hospital Târgu Mureș, 540136 Târgu Mureș, Romania
| | - Rodica Bălașa
- Ist Neurology Clinic, Emergency Clinical County Hospital Târgu Mureș, 540136 Târgu Mureș, Romania
- Department of Neurology, ‘George Emil Palade’ University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540136 Târgu Mureș, Romania
- Doctoral School, ‘George Emil Palade’ University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, 540142 Târgu Mureș, Romania
| |
Collapse
|
4
|
Xu W, Guo Y, Zhao L, Fu R, Qin X, Zhang Y, Cheng X, Xu S. The Aging Immune System: A Critical Attack on Ischemic Stroke. Mol Neurobiol 2025; 62:3322-3342. [PMID: 39271626 DOI: 10.1007/s12035-024-04464-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 08/29/2024] [Indexed: 09/15/2024]
Abstract
Ischemic stroke caused by cerebrovascular embolism is an age-related disease with high rates of disability and mortality. Although the mechanisms of immune and inflammatory development after stroke have been of great interest, most studies have neglected the critical and unavoidable factor of age. As the global aging trend intensifies, the number of stroke patients is constantly increasing, emphasizing the urgency of finding effective measures to address the needs of elderly stroke patients. The concept of "immunosenescence" appears to explain the worse stroke outcomes in older individuals. Immune remodeling due to aging involves dynamic changes at all levels of the immune system, and the overall consequences of central (brain-resident) and peripheral (non-brain-resident) immune cells in stroke vary according to the age of the individual. Lastly, the review outlines recent strategies aimed at immunosenescence to improve stroke prognosis.
Collapse
Affiliation(s)
- Wenzhe Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaoli Qin
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xueqi Cheng
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
5
|
Lei SY, Qu Y, Yang YQ, Liu JC, Zhang YF, Zhou SY, He QY, Jin H, Yang Y, Guo ZN. Cellular senescence: A novel therapeutic target for central nervous system diseases. Biomed Pharmacother 2024; 179:117311. [PMID: 39182322 DOI: 10.1016/j.biopha.2024.117311] [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: 05/27/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/27/2024] Open
Abstract
The underlying mechanisms of diseases affecting the central nervous system (CNS) remain unclear, limiting the development of effective therapeutic strategies. Remarkably, cellular senescence, a biological phenomenon observed in cultured fibroblasts in vitro, is a crucial intrinsic mechanism that influences homeostasis of the brain microenvironment and contributes to the onset and progression of CNS diseases. Cellular senescence has been observed in disease models established in vitro and in vivo and in bodily fluids or tissue components from patients with CNS diseases. These findings highlight cellular senescence as a promising target for preventing and treating CNS diseases. Consequently, emerging novel therapies targeting senescent cells have exhibited promising therapeutic effects in preclinical and clinical studies on aging-related diseases. These innovative therapies can potentially delay brain cell loss and functional changes, improve the prognosis of CNS diseases, and provide alternative treatments for patients. In this study, we examined the relevant advancements in this field, particularly focusing on the targeting of senescent cells in the brain for the treatment of chronic neurodegenerative diseases (e.g., Alzheimer's disease, Parkinson's disease, and multiple sclerosis) and acute neurotraumatic insults (e.g., ischemic stroke, spinal cord injury, and traumatic brain injury).
Collapse
Affiliation(s)
- Shuang-Yin Lei
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Yang Qu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Yu-Qian Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Jia-Cheng Liu
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Yi-Fei Zhang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Sheng-Yu Zhou
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Qian-Yan He
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China
| | - Hang Jin
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Yi Yang
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China.
| | - Zhen-Ni Guo
- Stroke Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China; Neuroscience Research Center, Department of Neurology, the First Hospital of Jilin University, Changchun 130021, China.
| |
Collapse
|
6
|
Wang G, Li Z, Wang G, Sun Q, Lin P, Wang Q, Zhang H, Wang Y, Zhang T, Cui F, Zhong Z. Advances in Engineered Nanoparticles for the Treatment of Ischemic Stroke by Enhancing Angiogenesis. Int J Nanomedicine 2024; 19:4377-4409. [PMID: 38774029 PMCID: PMC11108071 DOI: 10.2147/ijn.s463333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/02/2024] [Indexed: 05/24/2024] Open
Abstract
Angiogenesis, or the formation of new blood vessels, is a natural defensive mechanism that aids in the restoration of oxygen and nutrition delivery to injured brain tissue after an ischemic stroke. Angiogenesis, by increasing vessel development, may maintain brain perfusion, enabling neuronal survival, brain plasticity, and neurologic recovery. Induction of angiogenesis and the formation of new vessels aid in neurorepair processes such as neurogenesis and synaptogenesis. Advanced nano drug delivery systems hold promise for treatment stroke by facilitating efficient transportation across the the blood-brain barrier and maintaining optimal drug concentrations. Nanoparticle has recently been shown to greatly boost angiogenesis and decrease vascular permeability, as well as improve neuroplasticity and neurological recovery after ischemic stroke. We describe current breakthroughs in the development of nanoparticle-based treatments for better angiogenesis therapy for ischemic stroke employing polymeric nanoparticles, liposomes, inorganic nanoparticles, and biomimetic nanoparticles in this study. We outline new nanoparticles in detail, review the hurdles and strategies for conveying nanoparticle to lesions, and demonstrate the most recent advances in nanoparticle in angiogenesis for stroke treatment.
Collapse
Affiliation(s)
- Guangtian Wang
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhihui Li
- Department of Neurology, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Gongchen Wang
- Department of Vascular Surgery, The Fourth Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, 150086, People’s Republic of China
| | - Qixu Sun
- Department of Gastroenterology, Penglai People’s Hospital, Yantai, Shandong, 265600, People’s Republic of China
| | - Peng Lin
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Qian Wang
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Huishu Zhang
- Teaching Center of Biotechnology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Yanyan Wang
- Teaching Center of Morphology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Tongshuai Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Feiyun Cui
- Department of Physiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| | - Zhaohua Zhong
- Teaching Center of Pathogenic Biology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
- Department of Microbiology, School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, 150081, People’s Republic of China
| |
Collapse
|
7
|
Zeng Y, Xue T, Zhang D, Lv M. Transcriptomic Analysis of lncRNAs and their mRNA Networks in Cerebral Ischemia in Young and Aged Mice. Comb Chem High Throughput Screen 2024; 27:823-833. [PMID: 37340753 DOI: 10.2174/1386207326666230619091603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 04/26/2023] [Accepted: 05/12/2023] [Indexed: 06/22/2023]
Abstract
BACKGROUND Ischemic stroke comprises 75% of all strokes and it is associated with a great frailty and casualty rate. Certain data suggest multiple long non-coding Ribonucleic Acids (lncRNAs) assist the transcriptional, post-transcriptional, and epigenetic regulation of genes expressed in the CNS (Central Nervous System). However, these studies generally focus on differences in the expression patterns of lncRNAs and Messenger Ribonucleic Acids (mRNAs) in tissue samples before and after cerebral ischemic injury, ignoring the effects of age. METHODS In this study, differentially expressed lncRNA analysis was performed based on RNAseq data from the transcriptomic analysis of murine brain microglia related to cerebral ischemia injury in mice at different ages (10 weeks and 18 months). RESULTS The results showed that the number of downregulate differentially expressed genes (DEGs) in aged mice was 37 less than in young mice. Among them, lncRNA Gm-15987, RP24- 80F7.5, XLOC_379730, XLOC_379726 were significantly down-regulated. Then, Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis indicated that these specific lncRNAs were mainly related to inflammation. Based on the lncRNA/mRNA coexpression network, the mRNA co-expressed with lncRNA was mainly enriched in pathways, such as immune system progression, immune response, cell adhesion, B cell activation, and T cell differentiation. Our results indicate that the downregulation of lncRNA, such as Gm-15987, RP24- 80F7.5, XLOC_379730, and XLOC_379726 in aged mice may attenuate microglial-induced inflammation via the progress of immune system progression immune response, cell adhesion, B cell activation, and T cell differentiation. CONCLUSION The reported lncRNAs and their target mRNA during this pathology have potentially key regulatory functions in the cerebral ischemia in aged mice while being important for diagnosing and treating cerebral ischemia in the elderly.
Collapse
Affiliation(s)
- Yuanyuan Zeng
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Tengteng Xue
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| | - Dayong Zhang
- Department of New Media and Arts, Harbin Institute of Technology, Harbin, 150001, China
| | - Manhua Lv
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang Province, China
| |
Collapse
|
8
|
Darban YM, Askari H, Ghasemi-Kasman M, Yavarpour-Bali H, Dehpanah A, Gholizade P, Nosratiyan N. The Role of Induced Pluripotent Stem Cells in the Treatment of Stroke. Curr Neuropharmacol 2024; 22:2368-2383. [PMID: 39403058 PMCID: PMC11451314 DOI: 10.2174/1570159x22666240603084558] [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: 10/01/2023] [Revised: 01/14/2024] [Accepted: 01/16/2024] [Indexed: 10/19/2024] Open
Abstract
Stroke is a neurological disorder with high disability and mortality rates. Almost 80% of stroke cases are ischemic stroke, and the remaining are hemorrhagic stroke. The only approved treatment for ischemic stroke is thrombolysis and/or thrombectomy. However, these treatments cannot sufficiently relieve the disease outcome, and many patients remain disabled even after effective thrombolysis. Therefore, rehabilitative therapies are necessary to induce remodeling in the brain. Currently, stem cell transplantation, especially via the use of induced pluripotent stem cells (iPSCs), is considered a promising alternative therapy for stimulating neurogenesis and brain remodeling. iPSCs are generated from somatic cells by specific transcription factors. The biological functions of iPSCs are similar to those of embryonic stem cells (ESCs), including immunomodulation, reduced cerebral blood flow, cerebral edema, and autophagy. Although iPSC therapy plays a promising role in both hemorrhagic and ischemic stroke, its application is associated with certain limitations. Tumor formation, immune rejection, stem cell survival, and migration are some concerns associated with stem cell therapy. Therefore, cell-free therapy as an alternative method can overcome these limitations. This study reviews the therapeutic application of iPSCs in stroke models and the underlying mechanisms and constraints of these cells. Moreover, cell-free therapy using exosomes, apoptotic bodies, and microvesicles as alternative treatments is discussed.
Collapse
Affiliation(s)
| | - Hamid Askari
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Maryam Ghasemi-Kasman
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran
- Department of Physiology, School of Medicine, Babol University of Medical Sciences, Babol, Iran
| | | | - Amirabbas Dehpanah
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Parnia Gholizade
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| | - Nasrin Nosratiyan
- Student Research Committee, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
9
|
Wei H, Zhen L, Wang S, Yang L, Zhang S, Zhang Y, Jia P, Wang T, Wang K, Zhang Y, Ma L, Lv J, Zhang P. Glyceryl triacetate promotes blood-brain barrier recovery after ischemic stroke through lipogenesis-mediated IL-33 in mice. J Neuroinflammation 2023; 20:264. [PMID: 37968698 PMCID: PMC10648711 DOI: 10.1186/s12974-023-02942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Accepted: 10/30/2023] [Indexed: 11/17/2023] Open
Abstract
BACKGROUND Lipid metabolism has a crucial role in neural repair in neurodegenerative diseases. We recently revealed that lipogenesis-mediated interleukin-33 (IL-33) upregulation lead to blood-brain barrier (BBB) repair after ischemic stroke. However, manipulating the key enzyme fatty acid synthase (FASN) to enhance lipogenesis was very challenging. Glyceryl triacetate (GTA) was used as a donor of acetate and precursor of acetyl coenzyme A, the key substrate for de novo lipogenesis catalyzed by FASN. Therefore, we hypothesized that GTA would promote lipogenesis the peri-infarct after ischemic stroke and contribute to the BBB repair through IL-33. METHODS Middle cerebral artery occlusion (MCAO) was performed on C57BL mice and GTA was gavage administrated (4 g/kg) on day 2 and 4 after MCAO. Lipogenesis was evaluated by assessment of the protein level of FASN, lipid droplets, and fatty acid products through liquid chromatography-mass spectrometry in the peri-infarct area on day 3 after MCAO, respectively. BBB permeability was determined by extravasation of Evans blue, IgG and dextran, and levels of tight junction proteins in the peri-infarct area on day 7 after MCAO, respectively. Infarct size and neurological defects were assessed on day 7 after MCAO. Brain atrophy on day 30 and long-term sensorimotor abilities after MCAO were analyzed as well. The inhibitor of FASN, C75 and the virus-delivered FASN shRNA were used to evaluate the role of FASN-driven lipogenesis in GTA-improved BBB repair. Finally, the therapeutic potential of recombinant IL-33 on BBB repair and neurological recovery was evaluated. RESULTS We found that treatment with GTA increased the lipogenesis as evidenced by lipid droplets level and lauric acid content, but not the FASN protein level. Treatment with GTA increased the IL-33 level in the peri-infarct area and decreased the BBB permeability after MCAO. However, infarct size and neurological defect score were unchanged on day 7 after MCAO, while the long-term recovery of sensorimotor function and brain atrophy were improved by GTA. Inhibition of lipogenesis using C75 or FASN shRNA reversed the beneficial effect of GTA. Finally, exogenous IL-33 improved BBB repair and long-term functional recovery after stroke. CONCLUSION Collectively, we concluded that treatment with GTA improved the BBB repair and functional recovery after ischemic stroke, probably by the enhancement of lipogenesis and IL-33 expression.
Collapse
Affiliation(s)
- Haidong Wei
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Luming Zhen
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Liufei Yang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Shuyue Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Yuanyuan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Pengyu Jia
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Tianyue Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Kui Wang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Yan Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Lei Ma
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Jianrui Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China
| | - Pengbo Zhang
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi, China.
| |
Collapse
|
10
|
He X, Yu H, Fang J, Qi Z, Pei S, Yan B, Liu R, Wang Q, Szeto IMY, Liu B, Chen L, Li D. The effect of n-3 polyunsaturated fatty acid supplementation on cognitive function outcomes in the elderly depends on the baseline omega-3 index. Food Funct 2023; 14:9506-9517. [PMID: 37840364 DOI: 10.1039/d3fo02959j] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2023]
Abstract
Both epidemiological and preclinical studies have shown the benefits of n-3 polyunsaturated fatty acid (n-3 PUFA) on dementia and cognitive impairment, yet the results of clinical randomized controlled trials (RCTs) performed to date are conflicting. The difference in the baseline omega-3 index (O3i) of subjects is a potential cause for this disparity, yet this is usually ignored. The present meta-analysis aimed to evaluate the effect of n-3 polyunsaturated fatty acid (n-3 PUFA) on cognitive function in the elderly and the role of baseline O3i. A systematic literature search was conducted in PubMed, Embase, Cochrane Library, and Web of Science up to June 27th, 2023. The mean changes in the mini-mental state examination (MMSE) score were calculated as weighted mean differences by using a fixed-effects model. Fifteen random controlled trials were included in the meta-analysis. Pooled analysis showed that n-3 PUFA supplementation did not significantly improve the MMSE score (WMD = 0.04, [-0.08, 0.16]; Z = 0.62, P = 0.53; I2 = 0.00%, P(I2) = 0.49). Out of the 15 studies included in the meta-analysis, only 7 reported O3i at baseline and outcome, so only these 7 articles were used for subgroup analysis. Subgroup analysis showed that the MMSE score was significantly improved in the higher baseline O3i subgroup (WMD = 0.553, [0.01, 1.095]; I2 = 0.00%, P(I2) = 0.556) and higher O3i increment subgroup (WMD = 0.525, [0.023, 1.026]; I2 = 0.00%, P(I2) = 0.545). The overall effect demonstrated that n-3 PUFA supplementation exerted no improvement on global cognitive function. However, a higher baseline O3i and higher O3i increment were associated with an improvement in cognitive function in the elderly.
Collapse
Affiliation(s)
- Xin He
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- School of Public Health and Emergency Management, Southern University of Science and Technology, Shenzhen, China
| | - Hongzhuan Yu
- Weifang Traditional Chinese Hospital, Weifang, China
| | - Jiacheng Fang
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | - Zhongshi Qi
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | - Shengjie Pei
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | - Bei Yan
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | - Run Liu
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | - Qiuzhen Wang
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | | | - Biao Liu
- National Center of Technology Innovation for Dairy, Hohhot 010110, China
| | - Lei Chen
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
| | - Duo Li
- Institute of Nutrition & Health, Qingdao University, Qingdao, China.
- Department of Food Science and Nutrition, Zhejiang University, China
- Department of Nutrition, Dietetics and Food, Monash University, Australia
| |
Collapse
|
11
|
Zhang Y, Zhao X, Guo C, Zhang Y, Zeng F, Yin Q, Li Z, Shao L, Zhou D, Liu L. The Circadian System Is Essential for the Crosstalk of VEGF-Notch-mediated Endothelial Angiogenesis in Ischemic Stroke. Neurosci Bull 2023; 39:1375-1395. [PMID: 36862341 PMCID: PMC10465432 DOI: 10.1007/s12264-023-01042-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/18/2022] [Indexed: 03/03/2023] Open
Abstract
Ischemic stroke is a major public health problem worldwide. Although the circadian clock is involved in the process of ischemic stroke, the exact mechanism of the circadian clock in regulating angiogenesis after cerebral infarction remains unclear. In the present study, we determined that environmental circadian disruption (ECD) increased the stroke severity and impaired angiogenesis in the rat middle cerebral artery occlusion model, by measuring the infarct volume, neurological tests, and angiogenesis-related protein. We further report that Bmal1 plays an irreplaceable role in angiogenesis. Overexpression of Bmal1 promoted tube-forming, migration, and wound healing, and upregulated the vascular endothelial growth factor (VEGF) and Notch pathway protein levels. This promoting effect was reversed by the Notch pathway inhibitor DAPT, according to the results of angiogenesis capacity and VEGF pathway protein level. In conclusion, our study reveals the intervention of ECD in angiogenesis in ischemic stroke and further identifies the exact mechanism by which Bmal1 regulates angiogenesis through the VEGF-Notch1 pathway.
Collapse
Affiliation(s)
- Yuxing Zhang
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, 410208, China
- Hunan University of Chinese Medicine, Changsha, 410006, China
| | - Xin Zhao
- Hunan University of Chinese Medicine, Changsha, 410006, China
| | - Chun Guo
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Ying Zhang
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- Hunan University of Chinese Medicine, Changsha, 410006, China
| | - Fukang Zeng
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-Cerebral Diseases, Changsha, 410208, China
- Hunan University of Chinese Medicine, Changsha, 410006, China
| | - Qian Yin
- Hunan University of Chinese Medicine, Changsha, 410006, China
| | - Zhong Li
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China
| | - Le Shao
- Hunan University of Chinese Medicine, Changsha, 410006, China
- Laboratory of Prevention and Transformation of Major Diseases in Internal Medicine of Traditional Chinese Medicine, Changsha, 410007, China
| | - Desheng Zhou
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China.
| | - Lijuan Liu
- Department of Neurology, The First Hospital of Hunan University of Chinese Medicine, Changsha, 410007, China.
| |
Collapse
|
12
|
Chen T, Xia Y, Zhang L, Xu T, Yi Y, Chen J, Liu Z, Yang L, Chen S, Zhou X, Chen X, Wu H, Liu J. Loading neural stem cells on hydrogel scaffold improves cell retention rate and promotes functional recovery in traumatic brain injury. Mater Today Bio 2023; 19:100606. [PMID: 37063247 PMCID: PMC10102240 DOI: 10.1016/j.mtbio.2023.100606] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/23/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
Neural stem cell (NSC) has gained considerable attention in traumatic brain injury (TBI) treatment because of their ability to replenish dysfunctional neurons and stimulate endogenous neurorestorative processes. However, their therapeutic effects are hindered by the low cell retention rate after transplantation into the dynamic brain. In this study, we found cerebrospinal fluid (CSF) flow after TBI is an important factor associated with cell loss following NSC transplantation. Recently, several studies have shown that hydrogels could serve as a beneficial carrier for stem cell transplantation, which provides a solution to prevent CSF flow-induced cell loss after TBI. For this purpose, we evaluated three different hydrogel scaffolds and found the gelatin methacrylate (GelMA)/sodium alginate (Alg) (GelMA/Alg) hydrogel scaffold showed the best capabilities for NSC adherence, growth, and differentiation. Additionally, we detected that pre-differentiated NSCs, which were loaded on the GelMA/Alg hydrogel and cultured for 7 days in neuronal differentiation medium (NSC [7d]), had the highest cell retention rate after CSF impact. Next, the neuroprotective effects of the NSC-loaded GelMA/Alg hydrogel scaffold were evaluated in a rat model of TBI. NSC [7d]-loaded GelMA/Alg markedly decreased microglial activation and neuronal death in the acute phase, reduced tissue loss, alleviated astrogliosis, promoted neurogenesis, and improved neurological recovery in the chronic phase. In summary, we demonstrated that the integration with the GelMA/Alg and modification of NSC differentiation could inhibit the influence of CSF flow on transplanted NSCs, leading to increased number of retained NSCs and improved neuroprotective effects, providing a promising alternative for TBI treatment.
Collapse
Affiliation(s)
- Tiange Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuguo Xia
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liyang Zhang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Tao Xu
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, China
| | - Yan Yi
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Hunan, China
| | - Jianwei Chen
- Bio-Intelligent Manufacturing and Living Matter Bioprinting Center, Research Institute of Tsinghua University in Shenzhen, Tsinghua University, Shenzhen, China
| | - Ziyuan Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Liting Yang
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hypothalamic-Pituitary Research Center, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Siming Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoxi Zhou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xin Chen
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiyu Wu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinfang Liu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Corresponding author. Department of Neurosurgery, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, No. 87 Xiangya Rd, Kaifu District, Changsha, 410008, PR China.
| |
Collapse
|
13
|
Li Q, Niu X, Yi Y, Chen Y, Yuan J, Zhang J, Li H, Xia Y, Wang Y, Deng Z. Inducible Pluripotent Stem Cell-Derived Small Extracellular Vesicles Rejuvenate Senescent Blood-Brain Barrier to Protect against Ischemic Stroke in Aged Mice. ACS NANO 2023; 17:775-789. [PMID: 36562422 DOI: 10.1021/acsnano.2c10824] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Blood-brain barrier (BBB) breakdown after ischemic stroke exacerbates brain injury and BBB senescence can cause severe neurological deficits in aged ischemic stroke population. Recent evidence reveals that inducible pluripotent stem cell-derived small extracellular vesicles (iPSC-sEVs) possess phenomenal antisenescence capability. However, whether iPSC-sEVs can rejuvenate BBB senescence to improve stroke outcomes in aged mice remains unknown. Here, we showed that long-term treatment with iPSC-sEVs alleviated aging-induced BBB senescence in aged mice. In aged stroke mice, iPSC-sEVs significantly mitigated BBB integrity damage, reduced the following infiltration of peripheral leukocytes, and decreased the release of pro-inflammatory factors from the leukocytes, which ultimately inhibited neuronal death and improved neurofunctional recovery. Mechanism studies showed that iPSC-sEVs could activate the endothelial nitric oxide synthase (eNOS) and up-regulate sirtuin 1 (Sirt1) in senescent endothelial cells. Blocking the activation of eNOS abolished iPSC-sEV-mediated rejuvenation of BBB senescence and the protection of BBB integrity. Proteomics results demonstrated that iPSC-sEVs were enriched with bioactive factors including AKT serine/threonine kinase 1 (AKT1) and calmodulin (CALM) to activate the eNOS-Sirt1 axis. Further investigation showed that AKT1 and CALM inhibitors blocked iPSC-sEV-afforded activation of the eNOS-Sirt1 axis in senescent endothelial cells. Taken together, iPSC-sEVs can protect against ischemic stroke in aged mice by rejuvenating BBB senescence, partially, through delivering AKT1 and CALM to activate eNOS-Sirt1 axis, which indicates that iPSC-sEVs treatment is an effective alternative to treat ischemic stroke in the aged population.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Haiyan Li
- Chemical and Environment Engineering Department, School of Engineering, RMIT University, Melbourne, VIC 3001, Australia
| | | | | | | |
Collapse
|
14
|
Wu Y, Zhang J, Feng X, Jiao W. Omega-3 polyunsaturated fatty acids alleviate early brain injury after traumatic brain injury by inhibiting neuroinflammation and necroptosis. Transl Neurosci 2023; 14:20220277. [PMID: 36895263 PMCID: PMC9990778 DOI: 10.1515/tnsci-2022-0277] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 02/10/2023] [Indexed: 03/08/2023] Open
Abstract
Presently, traumatic brain injury (TBI) is a leading contributor to disability and mortality that places a considerable financial burden on countries all over the world. Docosahexaenoic acid and eicosapentaenoic acid are two kinds of omega-3 polyunsaturated fatty acids (ω-3 PUFA), both of which have been shown to have beneficial biologically active anti-inflammatory and antioxidant effects. However, the neuroprotective effect of ω-3 PUFA in TBI has not been proven, and its probable mechanism remains obscure. We suppose that ω-3 PUFA can alleviate early brain injury (EBI) via regulating necroptosis and neuroinflammation after TBI. This research intended to examine the neuroprotective effect of ω-3 and its possible molecular pathways in a C57BL/6 mice model of EBI caused by TBI. Cognitive function was assessed by measuring the neuronal necroptosis, neuroinflammatory cytokine levels, brain water content, and neurological score. The findings demonstrate that administration of ω-3 remarkably elevated neurological scores, alleviated cerebral edema, and reduced inflammatory cytokine levels of NF-κB, interleukin-1β (IL-1β), IL-6, and TNF-α, illustrating that ω-3 PUFA attenuated neuroinflammation, necroptosis, and neuronal cell death following TBI. The PPARγ/NF-κB signaling pathway is partially responsible for the neuroprotective activity of ω-3. Collectively, our findings illustrate that ω-3 can alleviate EBI after TBI against neuroinflammation and necroptosis.
Collapse
Affiliation(s)
- Yali Wu
- Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, 214044, China
| | - Jing Zhang
- Department of Neurosurgery, The Fourth People's Hospital of Taizhou, Taizhou, 225300, China
| | - Xiaoyan Feng
- Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi, 214044, China
| | - Wei Jiao
- Department of Nursing, 904th Hospital of Joint Logistic Support Force of PLA, 101 Xing Yuan North Road, Wuxi, 214044, China
| |
Collapse
|
15
|
Ha GH, Kim EJ, Park JS, Kim JE, Nam H, Yeon JY, Lee SH, Lee K, Kim CK, Joo KM. JAK2/STAT3 pathway mediates neuroprotective and pro-angiogenic treatment effects of adult human neural stem cells in middle cerebral artery occlusion stroke animal models. Aging (Albany NY) 2022; 14:8944-8969. [PMID: 36446389 PMCID: PMC9740376 DOI: 10.18632/aging.204410] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 11/17/2022] [Indexed: 12/03/2022]
Abstract
Mismatches between pre-clinical and clinical results of stem cell therapeutics for ischemic stroke limit their clinical applicability. To overcome these discrepancies, precise planning of pre-clinical experiments that can be translated to clinical trials and the scientific elucidation of treatment mechanisms is important. In this study, adult human neural stem cells (ahNSCs) derived from temporal lobe surgical samples were used (to avoid ethical and safety issues), and their therapeutic effects on ischemic stroke were examined using middle cerebral artery occlusion animal models. 5 × 105 ahNSCs was directly injected into the lateral ventricle of contralateral brain hemispheres of immune suppressed rat stroke models at the subacute phase of stroke. Compared with the mock-treated group, ahNSCs reduced brain tissue atrophy and neurological sensorimotor and memory functional loss. Tissue analysis demonstrated that the significant therapeutic effects were mediated by the neuroprotective and pro-angiogenic activities of ahNSCs, which preserved neurons in ischemic brain areas and decreased reactive astrogliosis and microglial activation. The neuroprotective and pro-angiogenic effects of ahNSCs were validated in in vitro stroke models and were induced by paracrine factors excreted by ahNSCs. When the JAK2/STAT3 signaling pathway was inhibited by a specific inhibitor, AG490, the paracrine neuroprotective and pro-angiogenic effects of ahNSCs were reversed. This pre-clinical study that closely simulated clinical settings and provided treatment mechanisms of ahNSCs for ischemic stroke may aid the development of protocols for subsequent clinical trials of ahNSCs and the realization of clinically available stem cell therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Geun-Hyoung Ha
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea
| | - Eun Ji Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea
| | - Jee Soo Park
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea
| | - Ji Eun Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea
| | - Hyun Nam
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, South Korea,Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Je Young Yeon
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea
| | - Sun-Ho Lee
- Department of Neurosurgery, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, South Korea,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, South Korea
| | - Kyunghoon Lee
- Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, South Korea,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| | - Chung Kwon Kim
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| | - Kyeung Min Joo
- Medical Innovation Technology Inc. (MEDINNO Inc.), Seoul 08513, South Korea,Department of Anatomy and Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, South Korea,Stem Cell and Regenerative Medicine Center, Research Institute for Future Medicine, Samsung Medical Center, Seoul 06351, South Korea,Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, Seoul 06351, South Korea,Single Cell Network Research Center, Sungkyunkwan University School of Medicine, Suwon 16149, South Korea,Biomedical Institute for Convergence at SKKU (BICS), Sungkyunkwan University, Suwon 16419, South Korea
| |
Collapse
|
16
|
Liu L, Liu J, Li M, Lyu J, Su W, Feng S, Ji X. Selective brain hypothermia attenuates focal cerebral ischemic injury and improves long-term neurological outcome in aged female mice. CNS Neurosci Ther 2022; 29:129-139. [PMID: 36341958 PMCID: PMC9804044 DOI: 10.1111/cns.14017] [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: 06/24/2022] [Revised: 10/09/2022] [Accepted: 10/20/2022] [Indexed: 11/09/2022] Open
Abstract
AIMS This study aimed to investigate the effects of mild selective brain hypothermia on aged female ischemic mice. METHODS A distal middle cerebral artery occlusion (dMCAO) model was established in aged female mice, who were then subjected to mild selective brain hypothermia immediately after the dMCAO procedure. Neurological behavioral examinations were conducted prior to and up to 35 days post-ischemia. Infarct volume, brain atrophy, pro-inflammation, and anti-inflammation microglia/macrophages phenotype and white matter injury were evaluated by immunofluorescence staining. Correlations between neurological behaviors and histological parameters were evaluated by Pearson product linear regression analysis. RESULTS Sensorimotor and cognitive function tests confirmed the protective effect of mild selective brain hypothermia in elderly female ischemic mice. In addition, hypothermia decreased the infarct volume and brain atrophy induced by focal cerebral ischemia. Furthermore, hypothermia alleviated ischemia-induced short-term and long-term white matter injury, which was correlated with behavioral deficits. Finally, hypothermia suppressed the harmful immunological response by promoting the transformation of pro-inflammatory microglia/macrophages to anti-inflammatory phenotype. This polarization was negatively correlated with neuronal loss and white matter injury. CONCLUSION Mild selective brain hypothermia promoted long-term functional recovery by alleviating white matter damage in an aged female mouse model of ischemia.
Collapse
Affiliation(s)
- Liqiang Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Jia Liu
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Ming Li
- Beijing Institute of Geriatrics, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Junxuan Lyu
- Department of Neurology, Xuanwu HospitalCapital Medical UniversityBeijingChina
| | - Wei Su
- Department of Neurosurgery, Beijing Tsing Hua Chang Gung Hospital, School of Clinical MedicineTsing Hua UniversityBeijingChina
| | - Shejun Feng
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina
| | - Xunming Ji
- Beijing Institute of Brain Disorders, Laboratory of Brain Disorders, Ministry of Science and Technology, Collaborative Innovation Center for Brain DisordersCapital Medical UniversityBeijingChina,Department of Neurosurgery, Xuanwu HospitalCapital Medical UniversityBeijingChina
| |
Collapse
|
17
|
Gao C, Nie M, Huang J, Tian Y, Wang D, Zhang J, Jiang R. Pharmacotherapy for mild traumatic brain injury: an overview of the current treatment options. Expert Opin Pharmacother 2022; 23:805-813. [PMID: 35290753 DOI: 10.1080/14656566.2022.2054328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Accounting for 90% of all traumatic brain injuries (TBIs), mild traumatic brain injury (mTBI) is currently the most frequently seen type of TBI. Although most patients can recover from mTBI, some may suffer from prolonged symptoms for months to years after injury. Growing evidence indicates that mTBI is associated with neurodegenerative diseases including dementias and Parkinson's disease (PD). Pharmacological interventions are necessary to address the symptoms and avoid the adverse consequences of mTBI. AREAS COVERED To provide an overview of the current treatment options, the authors herein review the potential drugs to reduce the secondary damage and symptom-targeted therapy as well as the ongoing clinical trials about pharmacotherapy for mTBI. EXPERT OPINION There has been no consensus on the pharmacotherapy for mTBI. Several candidates including n-3 PUFAs, melatonin, NAC and statins show potential benefits in lessening the secondary injury and improving neurological deficits in pre-clinic studies, which, however, still need further investigation in clinical trials. The current pharmacotherapy for mTBI is empirical in nature and mainly targets to mitigate the symptoms. Well-designed clinical trials are now warranted to provide high level evidence.
Collapse
Affiliation(s)
- Chuang Gao
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| | - Meng Nie
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| | - Jinhao Huang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| | - Ye Tian
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| | - Dong Wang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| | - Jianning Zhang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| | - Rongcai Jiang
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin 300052, China.,Key Laboratory of Post -Neuroinjury Neuro -repair and Regeneration in Central Nervous System, Tianjin Neurological Institute, Ministry of Education and Tianjin, China
| |
Collapse
|
18
|
Wei J, Wang Y, Yang S, Hao Z, Pan X, Ma A. Plasma chromogranin A levels are associated with acute ischemic stroke with anterior circulation large vessel occlusion. Nutr Metab Cardiovasc Dis 2022; 32:195-202. [PMID: 34893409 DOI: 10.1016/j.numecd.2021.09.030] [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: 02/08/2021] [Revised: 08/17/2021] [Accepted: 09/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND AND AIMS To investigate the relationship between chromogranin A (CgA) levels and acute ischemic stroke (AIS), especially anterior circulation large vessel occlusion (LVO). METHODS AND RESULTS 587 subjects were included in this study, including 205 AIS patients with anterior circulation LVO and 205 nonocclusive patients, as well as 177 healthy controls. On admission, plasma CgA levels were measured and neurological deficits were assessed by the NIH Stroke Scale. Outcomes were assessed by the modified Rankin Scale at 3 months. The predictive properties of CgA were evaluated by receiver operating characteristic (ROC) curve analysis. Binary logistic analysis assessed the association of CgA levels and AIS or anterior circulation LVO. AIS patients had lower CgA levels than health controls (p < 0.001). Anterior circulation LVO patients had lower CgA levels than nonocclusive patients (p < 0.001). The area under the ROC curve of plasma CgA levels in predicting anterior circulation LVO from AIS was 0.744 and the optimal cutoff value was 15.49 ng/mL with a Youden value of 0.332. Logistic analysis showed that CgA ≤15.49 ng/mL remained an independent risk factor for anterior circulation LVO after adjusting for related factors (OR = 6.519, 95% CI: 3.790-11.214, p < 0.001). CgA was an independent protective factor for mild stroke and good prognosis (p = 0.009, p = 0.005); however, the association disappeared after adjusting for occlusion (p = 0.768, p = 0.335). CONCLUSION CgA levels were lower in AIS patients, especially in anterior circulation LVO patients. Lower CgA levels are potential biomarker for anterior circulation LVO, and they may indicate good prognosis at 3 months in AIS.
Collapse
Affiliation(s)
- Jin Wei
- Department of Neurology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Yuan Wang
- Department of Neurology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Shaonan Yang
- Department of Neurology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Zhongnan Hao
- Department of Neurology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Xudong Pan
- Department of Neurology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266100, Shandong, China.
| | - Aijun Ma
- Department of Neurology, The Affiliated Hospital of Qingdao University, 59 Haier Road, Qingdao, 266100, Shandong, China.
| |
Collapse
|
19
|
Yongyue Z, Yang S, Li Z, Rongjin Z, Shumin W. Functional Brain Imaging Based on the Neurovascular Unit for Evaluating Neural Networks after Strok. ADVANCED ULTRASOUND IN DIAGNOSIS AND THERAPY 2022. [DOI: 10.37015/audt.2022.210033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
|
20
|
Sun P, Ma F, Xu Y, Zhou C, Stetler RA, Yin KJ. Genetic deletion of endothelial microRNA-15a/16-1 promotes cerebral angiogenesis and neurological recovery in ischemic stroke through Src signaling pathway. J Cereb Blood Flow Metab 2021; 41:2725-2742. [PMID: 33910400 PMCID: PMC8504951 DOI: 10.1177/0271678x211010351] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Cerebral angiogenesis is tightly controlled by specific microRNAs (miRs), including the miR-15a/16-1 cluster. Recently, we reported that endothelium-specific conditional knockout of the miR-15a/16-1 cluster (EC-miR-15a/16-1 cKO) promotes post-stroke angiogenesis and improves long-term neurological recovery by increasing protein levels of VEGFA, FGF2, and their respective receptors VEGFR2 and FGFR1. Herein, we further investigated the underlying signaling mechanism of these pro-angiogenic factors after ischemic stroke using a selective Src family inhibitor AZD0530. EC-miR-15a/16-1 cKO and age- and sex-matched wild-type littermate (WT) mice were subjected to 1 h middle cerebral artery occlusion (MCAO) and 28d reperfusion. AZD0530 was administered daily by oral gavage to both genotypes of mice 3-21d after MCAO. Compared to WT, AZD0530 administration exacerbated spatial cognitive impairments and brain atrophy in EC-miR-15a/16-1 cKO mice following MCAO. AZD0530 also attenuated long-term recovery of blood flow and inhibited the formation of new microvessels, including functional vessels with blood circulation, in the penumbra of stroked cKO mice. Moreover, AZD0530 blocked the Src signaling pathway by downregulating phospho-Src and its downstream mediators (p-Stat3, p-Akt, p-FAK, p-p44/42 MAPK, p-p38 MAPK) in post-ischemic brains. Collectively, our data demonstrated that endothelium-targeted deletion of the miR-15a/16-1 cluster promotes post-stroke angiogenesis and improves long-term neurological recovery via activating Src signaling pathway.
Collapse
Affiliation(s)
- Ping Sun
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Feifei Ma
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yang Xu
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Chao Zhou
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - R Anne Stetler
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders and Recovery, Department of Neurology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Mao XY, Yin XX, Guan QW, Xia QX, Yang N, Zhou HH, Liu ZQ, Jin WL. Dietary nutrition for neurological disease therapy: Current status and future directions. Pharmacol Ther 2021; 226:107861. [PMID: 33901506 DOI: 10.1016/j.pharmthera.2021.107861] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 04/02/2021] [Accepted: 04/06/2021] [Indexed: 02/06/2023]
Abstract
Adequate food intake and relative abundance of dietary nutrients have undisputed effects on the brain function. There is now substantial evidence that dietary nutrition aids in the prevention and remediation of neurologic symptoms in diverse pathological conditions. The newly described influences of dietary factors on the alterations of mitochondrial dysfunction, epigenetic modification and neuroinflammation are important mechanisms that are responsible for the action of nutrients on the brain health. In this review, we discuss the state of evidence supporting that distinct dietary interventions including dietary supplement and dietary restriction have the ability to tackle neurological disorders using Alzheimer's disease, Parkinson's disease, stroke, epilepsy, traumatic brain injury, amyotrophic lateral sclerosis, Huntington's disease and multiple sclerosis as examples. Additionally, it is also highlighting that diverse potential mechanisms such as metabolic control, epigenetic modification, neuroinflammation and gut-brain axis are of utmost importance for nutrient supply to the risk of neurologic condition and therapeutic response. Finally, we also highlight the novel concept that dietary nutrient intervention reshapes metabolism-epigenetics-immunity cycle to remediate brain dysfunction. Targeting metabolism-epigenetics-immunity network will delineate a new blueprint for combating neurological weaknesses.
Collapse
Affiliation(s)
- Xiao-Yuan Mao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| | - Xi-Xi Yin
- Department of Pediatrics, Xiangya Hospital, Central South University, Changsha, China
| | - Qi-Wen Guan
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Qin-Xuan Xia
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Nan Yang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Hong-Hao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China
| | - Zhao-Qian Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha 410008, PR China; Institute of Clinical Pharmacology, Central South University, Hunan Key Laboratory of Pharmacogenetics, 110 Xiangya Road, Changsha 410078, PR China; Engineering Research Center of Applied Technology of Pharmacogenomics, Ministry of Education, 110 Xiangya Road, Changsha 410078, PR China; National Clinical Research Center for Geriatric Disorders, 87 Xiangya Road, Changsha 410008, Hunan, PR China.
| | - Wei-Lin Jin
- Institute of Cancer Neuroscience, Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, The First Clinical Medical College of Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
22
|
Pergande MR, Amoroso VG, Nguyen TTA, Li W, Vice E, Park TJ, Cologna SM. PPARα and PPARγ Signaling Is Enhanced in the Brain of the Naked Mole-Rat, a Mammal that Shows Intrinsic Neuroprotection from Oxygen Deprivation. J Proteome Res 2021; 20:4258-4271. [PMID: 34351155 DOI: 10.1021/acs.jproteome.1c00131] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Naked mole-rats (NMRs) are a long-lived animal that do not develop age-related diseases including neurodegeneration and cancer. Additionally, NMRs have a profound ability to consume reactive oxygen species (ROS) and survive long periods of oxygen deprivation. Here, we evaluated the unique proteome across selected brain regions of NMRs at different ages. Compared to mice, we observed numerous differentially expressed proteins related to altered mitochondrial function in all brain regions, suggesting that the mitochondria in NMRs may have adapted to compensate for energy demands associated with living in a harsh, underground environment. Keeping in mind that ROS can induce polyunsaturated fatty acid peroxidation under periods of neuronal stress, we investigated docosahexaenoic acid (DHA) and arachidonic acid (AA) peroxidation under oxygen-deprived conditions and observed that NMRs undergo DHA and AA peroxidation to a far less extent compared to mice. Further, our proteomic analysis also suggested enhanced peroxisome proliferator-activated receptor (PPAR)-retinoid X receptor (RXR) activation in NMRs via the PPARα-RXR and PPARγ-RXR complexes. Correspondingly, we present several lines of evidence supporting PPAR activation, including increased eicosapetenoic and omega-3 docosapentaenoic acid, as well as an upregulation of fatty acid-binding protein 3 and 4, known transporters of omega-3 fatty acids and PPAR activators. These results suggest enhanced PPARα and PPARγ signaling as a potential, innate neuroprotective mechanism in NMRs.
Collapse
Affiliation(s)
- Melissa R Pergande
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Vince G Amoroso
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Thu T A Nguyen
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Wenping Li
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Emily Vice
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Thomas J Park
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, Illinois 60607, United States.,Laboratory for Integrative Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois at Chicago, Chicago, Illinois 60607, United States.,Laboratory for Integrative Neuroscience, University of Illinois at Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
23
|
The Role of Supplementation with Natural Compounds in Post-Stroke Patients. Int J Mol Sci 2021; 22:ijms22157893. [PMID: 34360658 PMCID: PMC8348438 DOI: 10.3390/ijms22157893] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/07/2021] [Accepted: 07/20/2021] [Indexed: 12/13/2022] Open
Abstract
Malnutrition is a serious problem in post-stroke patients. Importantly, it intensifies with hospitalization, and is related to both somatic and psychological reasons, as well as is associated with the insufficient knowledge of people who accompany the patient. Malnutrition is a negative prognostic factor, leading to a reduction in the quality of life. Moreover, this condition significantly extends hospitalization time, increases the frequency of treatment in intensive care units, and negatively affects the effectiveness of rehabilitation. Obtaining growing data on the therapeutic effectiveness of new compounds of natural origin is possible through the use of pharmacodynamic and analytical methods to assess their therapeutic properties. The proper supply of nutrients, as well as compounds of natural origin, is an important element of post-stroke therapy, due to their strong antioxidant, anti-inflammatory, neuroprotective and neuroplasticity enhancing properties. Taking the above into account, in this review we present the current state of knowledge on the benefits of using selected substances of natural origin in patients after cerebral stroke.
Collapse
|
24
|
Pu H, Zheng X, Jiang X, Mu H, Xu F, Zhu W, Ye Q, Jizhang Y, Hitchens TK, Shi Y, Hu X, Leak RK, Dixon CE, Bennett MV, Chen J. Interleukin-4 improves white matter integrity and functional recovery after murine traumatic brain injury via oligodendroglial PPARγ. J Cereb Blood Flow Metab 2021; 41:511-529. [PMID: 32757740 PMCID: PMC7922743 DOI: 10.1177/0271678x20941393] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Long-term neurological recovery after severe traumatic brain injury (TBI) is strongly linked to the repair and functional restoration of injured white matter. Emerging evidence suggests that the anti-inflammatory cytokine interleukin-4 (IL-4) plays an important role in promoting white matter integrity after cerebral ischemic injury. Here, we report that delayed intranasal delivery of nanoparticle-packed IL-4 boosted sensorimotor neurological recovery in a murine model of controlled cortical impact, as assessed by a battery of neurobehavioral tests for up to five weeks. Post-injury IL-4 treatment failed to reduce macroscopic brain lesions after TBI, but preserved the structural and functional integrity of white matter, at least in part through oligodendrogenesis. IL-4 directly facilitated the differentiation of oligodendrocyte progenitor cells (OPCs) into mature myelin-producing oligodendrocytes in primary cultures, an effect that was attenuated by selective PPARγ inhibition. IL-4 treatment after TBI in vivo also failed to stimulate oligodendrogenesis or improve white matter integrity in OPC-specific PPARγ conditional knockout (cKO) mice. Accordingly, IL-4-afforded improvements in sensorimotor neurological recovery after TBI were markedly impaired in the PPARγ cKO mice compared to wildtype controls. These results support IL-4 as a potential novel neurorestorative therapy to improve white matter functionality and mitigate the long-term neurological consequences of TBI.
Collapse
Affiliation(s)
- Hongjian Pu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xuan Zheng
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoyan Jiang
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Hongfeng Mu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Fei Xu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wen Zhu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Ye
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yunneng Jizhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - T Kevin Hitchens
- Animal Imaging Center, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA.,Department of Neurobiology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yejie Shi
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rehana K Leak
- Graduate School of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA, USA
| | - C Edward Dixon
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Department of Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
| | - Michael Vl Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jun Chen
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA.,Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
25
|
Sommer CJ, Schäbitz WR. Principles and requirements for stroke recovery science. J Cereb Blood Flow Metab 2021; 41:471-485. [PMID: 33175596 PMCID: PMC7907998 DOI: 10.1177/0271678x20970048] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 10/03/2020] [Accepted: 10/05/2020] [Indexed: 12/11/2022]
Abstract
The disappointing results in bench-to-bedside translation of neuroprotective strategies caused a certain shift in stroke research towards enhancing the endogenous recovery potential of the brain. One reason for this focus on recovery is the much wider time window for therapeutic interventions which is open for at least several months. Since recently two large clinical studies using d-amphetamine or fluoxetine, respectively, to enhance post-stroke neurological outcome failed again it is a good time for a critical reflection on principles and requirements for stroke recovery science. In principal, stroke recovery science deals with all events from the molecular up to the functional and behavioral level occurring after brain ischemia eventually ending up with any measurable improvement of various clinical parameters. A detailed knowledge of the spontaneously occurring post-ischemic regeneration processes is the indispensable prerequisite for any therapeutic approaches aiming to modify these responses to enhance post-stroke recovery. This review will briefly illuminate the molecular mechanisms of post-ischemic regeneration and the principle possibilities to foster post-stroke recovery. In this context, recent translational approaches are analyzed. Finally, the principal and specific requirements and pitfalls in stroke recovery research as well as potential explanations for translational failures will be discussed.
Collapse
Affiliation(s)
- Clemens J Sommer
- Institute of Neuropathology, University Medical Center of the
Johannes Gutenberg-University Mainz, Mainz, Germany
| | | |
Collapse
|
26
|
Role of polyunsaturated fatty acids in ischemic stroke - A perspective of specialized pro-resolving mediators. Clin Nutr 2021; 40:2974-2987. [PMID: 33509668 DOI: 10.1016/j.clnu.2020.12.037] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) have been proposed as beneficial for cardiovascular health. However, results from both epidemiological studies and clinical trials have been inconsistent, whereas most of the animal studies showed promising benefits of PUFAs in the prevention and treatment of ischemic stroke. In recent years, it has become clear that PUFAs are metabolized into various types of bioactive derivatives, including the specialized pro-resolving mediators (SPMs). SPMs exert multiple biofunctions, such as to limit excessive inflammatory responses, regulate lipid metabolism and immune cell functions, decrease production of pro-inflammatory factors, increase anti-inflammatory mediators, as well as to promote tissue repair and homeostasis. Inflammation has been recognised as a key contributor to the pathophysiology of acute ischemic stroke. Owing to their potent pro-resolving actions, SPMs are potential for development of novel anti-stroke therapy. In this review, we will summarize current knowledge of epidemiological studies, basic research and clinical trials concerning PUFAs in stroke prevention and treatment, with special attention to SPMs as the unsung heroes behind PUFAs.
Collapse
|
27
|
Candelario-Jalil E, Paul S. Impact of aging and comorbidities on ischemic stroke outcomes in preclinical animal models: A translational perspective. Exp Neurol 2021; 335:113494. [PMID: 33035516 PMCID: PMC7874968 DOI: 10.1016/j.expneurol.2020.113494] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Revised: 09/25/2020] [Accepted: 10/02/2020] [Indexed: 12/16/2022]
Abstract
Ischemic stroke is a highly complex and devastating neurological disease. The sudden loss of blood flow to a brain region due to an ischemic insult leads to severe damage to that area resulting in the formation of an infarcted tissue, also known as the ischemic core. This is surrounded by the peri-infarct region or penumbra that denotes the functionally impaired but potentially salvageable tissue. Thus, the penumbral tissue is the main target for the development of neuroprotective strategies to minimize the extent of ischemic brain damage by timely therapeutic intervention. Given the limitations of reperfusion therapies with recombinant tissue plasminogen activator or mechanical thrombectomy, there is high enthusiasm to combine reperfusion therapy with neuroprotective strategies to further reduce the progression of ischemic brain injury. Till date, a large number of candidate neuroprotective drugs have been identified as potential therapies based on highly promising results from studies in rodent ischemic stroke models. However, none of these interventions have shown therapeutic benefits in stroke patients in clinical trials. In this review article, we discussed the urgent need to utilize preclinical models of ischemic stroke that more accurately mimic the clinical conditions in stroke patients by incorporating aged animals and animal stroke models with comorbidities. We also outlined the recent findings that highlight the significant differences in stroke outcome between young and aged animals, and how major comorbid conditions such as hypertension, diabetes, obesity and hyperlipidemia dramatically increase the vulnerability of the brain to ischemic damage that eventually results in worse functional outcomes. It is evident from these earlier studies that including animal models of aging and comorbidities during the early stages of drug development could facilitate the identification of neuroprotective strategies with high likelihood of success in stroke clinical trials.
Collapse
Affiliation(s)
- Eduardo Candelario-Jalil
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32610, USA.
| | - Surojit Paul
- Department of Neurology, University of New Mexico Health Sciences Center, Albuquerque, NM 87131, USA
| |
Collapse
|
28
|
Shi L, Rocha M, Zhang W, Jiang M, Li S, Ye Q, Hassan SH, Liu L, Adair MN, Xu J, Luo J, Hu X, Wechsler LR, Chen J, Shi Y. Genome-wide transcriptomic analysis of microglia reveals impaired responses in aged mice after cerebral ischemia. J Cereb Blood Flow Metab 2020; 40:S49-S66. [PMID: 32438860 PMCID: PMC7687039 DOI: 10.1177/0271678x20925655] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 03/24/2020] [Accepted: 04/13/2020] [Indexed: 12/25/2022]
Abstract
Senescence-associated alterations in microglia may have profound impact on cerebral homeostasis and stroke outcomes. However, the lack of a transcriptome-wide comparison between young and aged microglia in the context of ischemia limits our understanding of aging-related mechanisms. Herein, we performed RNA sequencing analysis of microglia purified from cerebral hemispheres of young adult (10-week-old) and aged (18-month-old) mice five days after distal middle cerebral artery occlusion or after sham operation. Considerable transcriptional differences were observed between young and aged microglia in healthy brains, indicating heightened chronic inflammation in aged microglia. Following stroke, the overall transcriptional activation was more robust (>13-fold in the number of genes upregulated) in young microglia than in aged microglia. Gene clusters with functional implications in immune inflammatory responses, immune cell chemotaxis, tissue remodeling, and cell-cell interactions were markedly activated in microglia of young but not aged stroke mice. Consistent with the genomic profiling predictions, post-stroke cerebral infiltration of peripheral immune cells was markedly decreased in aged mice compared to young mice. Moreover, post-ischemic aged microglia demonstrated reduced interaction with neighboring neurons and diminished polarity toward the infarct lesion. These alterations in microglial gene response and behavior may contribute to aging-driven vulnerability and poorer recovery after ischemic stroke.
Collapse
Affiliation(s)
- Ligen Shi
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Marcelo Rocha
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Wenting Zhang
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ming Jiang
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sicheng Li
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Qing Ye
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Sulaiman H Hassan
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Liqiang Liu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Maya N Adair
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jing Xu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jianhua Luo
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoming Hu
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Lawrence R Wechsler
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Jun Chen
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| | - Yejie Shi
- Department of Neurology, Pittsburgh Institute of Brain Disorders & Recovery and UPMC Stroke Institute, University of Pittsburgh, Pittsburgh, PA, USA
- Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA, USA
| |
Collapse
|
29
|
Comhaire F, Decleer W. Can the biological mechanisms of ageing be corrected by food supplementation. The concept of health care over sick care. Aging Male 2020; 23:1146-1157. [PMID: 31973615 DOI: 10.1080/13685538.2020.1713080] [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] [Indexed: 10/25/2022] Open
Abstract
"From care for the sick to care for health" implies that age-related diseases and discomforts, which impair the quality of life, should be prevented rather than treated. Healthy lifestyle and nutrition, and hormone supplementation - when needed - are of crucial importance. Food supplementation with nutraceuticals composed of vitamins, oligo-minerals, plant extracts and essential amino- and fatty acids should reduce age-related oxidative and epigenetic damage to DNA, and inhibit inflammatory and metabolic impairment. This study of the potential beneficial effects of novel nutraceuticals on the biological mechanisms of physical and mental ageing suggests these supplements may be scientifically justified. In the absence of adverse side effects and the expected favourable effect on the quality-adjusted life years, the benefit over risk ratio of nutraceutical supplementation should be positive.
Collapse
Affiliation(s)
| | - Wim Decleer
- Department of Reproductive Medicine, AZ Palfijn, Ghent, Belgium
- Fertility Clinic, Aalter, Belgium
| |
Collapse
|
30
|
Yamagata K. Dietary docosahexaenoic acid inhibits neurodegeneration and prevents stroke. J Neurosci Res 2020; 99:561-572. [PMID: 32964457 DOI: 10.1002/jnr.24728] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 07/11/2020] [Accepted: 08/30/2020] [Indexed: 01/07/2023]
Abstract
Stroke severely impairs quality of life and has a high mortality rate. On the other hand, dietary docosahexaenoic acid (DHA) prevents neuronal damage. In this review, we describe the effects of dietary DHA on ischemic stroke-associated neuronal damage and its role in stroke prevention. Recent epidemiological studies have been conducted to analyze stroke prevention through DHA intake. The effects of dietary intake and supply of DHA to neuronal cells, DHA-mediated inhibition of neuronal damage, and its mechanism, including the effects of the DHA metabolite, neuroprotectin D1 (NPD1), were investigated. These studies revealed that DHA intake was associated with a reduced risk of stroke. Moreover, studies have shown that DHA intake may reduce stroke mortality rates. DHA, which is abundant in fish oil, passes through the blood-brain barrier to accumulate as a constituent of phospholipids in the cell membranes of neuronal cells and astrocytes. Astrocytes supply DHA to neuronal cells, and neuronal DHA, in turn, activates Akt and Raf-1 to prevent neuronal death or damage. Therefore, DHA indirectly prevents neuronal damage. Furthermore, NDP1 blocks neuronal apoptosis. DHA, together with NPD1, may block neuronal damage and prevent stroke. The inhibitory effect on neuronal damage is achieved through the antioxidant (via inducing the Nrf2/HO-1 system) and anti-inflammatory effects (via promoting JNK/AP-1 signaling) of DHA.
Collapse
Affiliation(s)
- Kazuo Yamagata
- Department of Food Bioscience & Biotechnology, College of Bioresource Science, Nihon University (UNBS), Fujisawa, Japan
| |
Collapse
|
31
|
Xia Y, Ling X, Hu G, Zhu Q, Zhang J, Li Q, Zhao B, Wang Y, Deng Z. Small extracellular vesicles secreted by human iPSC-derived MSC enhance angiogenesis through inhibiting STAT3-dependent autophagy in ischemic stroke. Stem Cell Res Ther 2020; 11:313. [PMID: 32698909 PMCID: PMC7374834 DOI: 10.1186/s13287-020-01834-0] [Citation(s) in RCA: 111] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/21/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
Background Small extracellular vesicles (sEV) secreted by mesenchymal stem cells (MSC) derived from human induced pluripotent stem cells (iPSC, iMSC-sEV) are considered to have great potential in treating ischemic diseases. Angiogenesis play an important role in post-stroke recovery. However, no studies have yet been conducted to systemically examine the effect and the underlying mechanism of iMSC-sEV on angiogenesis under brain ischemia conditions. Methods Ischemic stroke model was performed in rats induced by middle cerebral artery occlusion (MCAO), and the pro-angiogenic capacity of iMSC-sEV was measured. The in vitro effects of iMSC-sEV on the migration and tube formation of endothelial cells were investigated, respectively. Autophagy and autophagy-related signaling pathway were detected in vivo and in vitro. Results We found that iMSC-sEV significantly reduced infarct volume, enhanced angiogenesis, and alleviated long-term neurological deficits in rats after stroke. We also demonstrated that iMSC-sEV increased migration and tube formation of endothelial cells in vitro. A further mechanism study revealed that the pro-angiogenic effect of iMSC-sEV was correlated with a reduction in autophagy. Furthermore, iMSC-sEV significantly activated signal transducer and activator of transcription 3 (STAT3), and suppression of STAT3 abolished iMSC-sEV-induced inhibition of autophagy and promotion of angiogenesis in vivo and in vitro. Conclusions Taken together, our data indicate that iMSC-sEV promote angiogenesis after ischemic stroke, potentially, by inhibiting autophagy, a process that is partially dependent on STAT3 activation.
Collapse
Affiliation(s)
- Yuguo Xia
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Xiaozheng Ling
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Guowen Hu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Qingwei Zhu
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Juntao Zhang
- Institute of Microsurgery and Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Qing Li
- Institute of Microsurgery and Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Bizeng Zhao
- Institute of Microsurgery and Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China
| | - Yang Wang
- Institute of Microsurgery and Extremities, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| | - Zhifeng Deng
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, No. 600 Yishan Road, Shanghai, 200233, China.
| |
Collapse
|
32
|
Ren C, Li N, Gao C, Zhang W, Yang Y, Li S, Ji X, Ding Y. Ligustilide provides neuroprotection by promoting angiogenesis after cerebral ischemia. Neurol Res 2020; 42:683-692. [PMID: 32584207 DOI: 10.1080/01616412.2020.1782122] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Changhong Ren
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Ning Li
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Chen Gao
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Wei Zhang
- Department of Herbal Formula Science Medicine, Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Yong Yang
- Department of Herbal Formula Science Medicine, Chinese Medicine School, Beijing University of Chinese Medicine, Beijing, China
| | - Sijie Li
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
| | - Xunming Ji
- Institute of Hypoxia Medicine, Xuanwu Hospital, Capital Medical University, Beijing, China
- BeijingKey Laboratory of Hypoxia Translational Medicine, Beijing, China
- Center of Stroke, Beijing Institute for Brain Disorder, Beijing, China
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, MI, USA
| |
Collapse
|
33
|
Pacheva I, Ivanov I. Targeted Biomedical Treatment for Autism Spectrum Disorders. Curr Pharm Des 2020; 25:4430-4453. [PMID: 31801452 DOI: 10.2174/1381612825666191205091312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 12/02/2019] [Indexed: 12/29/2022]
Abstract
BACKGROUND A diagnosis of autism spectrum disorders (ASD) represents presentations with impairment in communication and behaviour that vary considerably in their clinical manifestations and etiology as well as in their likely pathophysiology. A growing body of data indicates that the deleterious effect of oxidative stress, mitochondrial dysfunction, immune dysregulation and neuroinflammation, as well as their interconnections are important aspects of the pathophysiology of ASD. Glutathione deficiency decreases the mitochondrial protection against oxidants and tumor necrosis factor (TNF)-α; immune dysregulation and inflammation inhibit mitochondrial function through TNF-α; autoantibodies against the folate receptors underpin cerebral folate deficiency, resulting in disturbed methylation, and mitochondrial dysfunction. Such pathophysiological processes can arise from environmental and epigenetic factors as well as their combined interactions, such as environmental toxicant exposures in individuals with (epi)genetically impaired detoxification. The emerging evidence on biochemical alterations in ASD is forming the basis for treatments aimed to target its biological underpinnings, which is of some importance, given the uncertain and slow effects of the various educational interventions most commonly used. METHODS Literature-based review of the biomedical treatment options for ASD that are derived from established pathophysiological processes. RESULTS Most proposed biomedical treatments show significant clinical utility only in ASD subgroups, with specified pre-treatment biomarkers that are ameliorated by the specified treatment. For example, folinic acid supplementation has positive effects in ASD patients with identified folate receptor autoantibodies, whilst the clinical utility of methylcobalamine is apparent in ASD patients with impaired methylation capacity. Mitochondrial modulating cofactors should be considered when mitochondrial dysfunction is evident, although further research is required to identify the most appropriate single or combined treatment. Multivitamins/multiminerals formulas, as well as biotin, seem appropriate following the identification of metabolic abnormalities, with doses tapered to individual requirements. A promising area, requiring further investigations, is the utilization of antipurinergic therapies, such as low dose suramin. CONCLUSION The assessment and identification of relevant physiological alterations and targeted intervention are more likely to produce positive treatment outcomes. As such, current evidence indicates the utility of an approach based on personalized and evidence-based medicine, rather than treatment targeted to all that may not always be beneficial (primum non nocere).
Collapse
Affiliation(s)
- Iliyana Pacheva
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| | - Ivan Ivanov
- Department of Pediatrics and Medical Genetics, Medical University - Plovdiv, Plovdiv 4002, Bulgaria
| |
Collapse
|
34
|
Sun P, Zhang K, Hassan SH, Zhang X, Tang X, Pu H, Stetler RA, Chen J, Yin KJ. Endothelium-Targeted Deletion of microRNA-15a/16-1 Promotes Poststroke Angiogenesis and Improves Long-Term Neurological Recovery. Circ Res 2020; 126:1040-1057. [PMID: 32131693 DOI: 10.1161/circresaha.119.315886] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
RATIONALE Angiogenesis promotes neurological recovery after stroke and is associated with longer survival of stroke patients. Cerebral angiogenesis is tightly controlled by certain microRNAs (miRs), such as the miR-15a/16-1 cluster, among others. However, the function of the miR-15a/16-1 cluster in endothelium on postischemic cerebral angiogenesis is not known. OBJECTIVE To investigate the functional significance and molecular mechanism of endothelial miR-15a/16-1 cluster on angiogenesis in the ischemic brain. METHODS AND RESULTS Endothelial cell-selective miR-15a/16-1 conditional knockout (EC-miR-15a/16-1 cKO) mice and wild-type littermate controls were subjected to 1 hour middle cerebral artery occlusion followed by 28-day reperfusion. Deletion of miR-15a/16-1 cluster in endothelium attenuates post-stroke brain infarction and atrophy and improves the long-term sensorimotor and cognitive recovery against ischemic stroke. Endothelium-targeted deletion of the miR-15a/16-1 cluster also enhances post-stroke angiogenesis by promoting vascular remodeling and stimulating the generation of newly formed functional vessels, and increases the ipsilateral cerebral blood flow. Endothelial cell-selective deletion of the miR-15a/16-1 cluster up-regulated the protein expression of pro-angiogenic factors VEGFA (vascular endothelial growth factor), FGF2 (fibroblast growth factor 2), and their receptors VEGFR2 (vascular endothelial growth factor receptor 2) and FGFR1 (fibroblast growth factor receptor 1) after ischemic stroke. Consistently, lentiviral knockdown of the miR-15a/16-1 cluster in primary mouse or human brain microvascular endothelial cell cultures enhanced in vitro angiogenesis and up-regulated pro-angiogenic proteins expression after oxygen-glucose deprivation, whereas lentiviral overexpression of the miR-15a/16-1 cluster suppressed in vitro angiogenesis and down-regulated pro-angiogenic proteins expression. Mechanistically, miR-15a/16-1 translationally represses pro-angiogenic factors VEGFA, FGF2, and their receptors VEGFR2 and FGFR1, respectively, by directly binding to the complementary sequences within 3'-untranslated regions of those messenger RNAs. CONCLUSIONS Endothelial miR-15a/16-1 cluster is a negative regulator for postischemic cerebral angiogenesis and long-term neurological recovery. Inhibition of miR-15a/16-1 function in cerebrovascular endothelium may be a legitimate therapeutic approach for stroke recovery.
Collapse
Affiliation(s)
- Ping Sun
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - Kai Zhang
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - Sulaiman H Hassan
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - Xuejing Zhang
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - Xuelian Tang
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - Hongjian Pu
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - R Anne Stetler
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.)
| | - Jun Chen
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.).,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, PA (J.C., K.-J.Y.)
| | - Ke-Jie Yin
- From the Department of Neurology, Pittsburgh Institute of Brain Disorders and Recovery, University of Pittsburgh School of Medicine, PA (P.S., K.Z., S.H.H., X.Z., X.T., H.P., R.A.S., J.C., K.-J.Y.).,Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Healthcare System, PA (J.C., K.-J.Y.)
| |
Collapse
|
35
|
Li Y, Xie L, Huang T, Zhang Y, Zhou J, Qi B, Wang X, Chen Z, Li P. Aging Neurovascular Unit and Potential Role of DNA Damage and Repair in Combating Vascular and Neurodegenerative Disorders. Front Neurosci 2019; 13:778. [PMID: 31440124 PMCID: PMC6694749 DOI: 10.3389/fnins.2019.00778] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2019] [Accepted: 07/11/2019] [Indexed: 02/01/2023] Open
Abstract
Progressive neurological deterioration poses enormous burden on the aging population with ischemic stroke and neurodegenerative disease patients, such as Alzheimers’ disease and Parkinson’s disease. The past two decades have witnessed remarkable advances in the research of neurovascular unit dysfunction, which is emerging as an important pathological feature that underlies these neurological disorders. Dysfunction of the unit allows penetration of blood-derived toxic proteins or leukocytes into the brain and contributes to white matter injury, disturbed neurovascular coupling and neuroinflammation, which all eventually lead to cognitive dysfunction. Recent evidences suggest that aging-related oxidative stress, accumulated DNA damage and impaired DNA repair capacities compromises the genome integrity not only in neurons, but also in other cell types of the neurovascular unit, such as endothelial cells, astrocytes and pericytes. Combating DNA damage or enhancing DNA repair capacities in the neurovascular unit represents a promising therapeutic strategy for vascular and neurodegenerative disorders. In this review, we focus on aging related mechanisms that underlie DNA damage and repair in the neurovascular unit and introduce several novel strategies that target the genome integrity in the neurovascular unit to combat the vascular and neurodegenerative disorders in the aging brain.
Collapse
Affiliation(s)
- Yan Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Lv Xie
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Tingting Huang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yueman Zhang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Zhou
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Bo Qi
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Wang
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Zengai Chen
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Peiying Li
- Department of Anesthesiology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
36
|
Baltan S, Shi Y, Keep RF, Chen J. The effect of aging on brain injury and recovery after stroke. Neurobiol Dis 2019; 126:1-2. [PMID: 31010535 DOI: 10.1016/j.nbd.2019.04.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- Selva Baltan
- Department of Neurosciences, Cleveland Clinic, Cleveland, OH 44195
| | - Yejie Shi
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh, PA 15261
| |
Collapse
|
37
|
Parikh M, Pierce GN. Dietary flaxseed: what we know and don't know about its effects on cardiovascular disease. Can J Physiol Pharmacol 2018; 97:75-81. [PMID: 30562057 DOI: 10.1139/cjpp-2018-0547] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Flaxseed (Linum usitatissimum) is composed of a unique combination of bioactive components that appear to generate, through either an isolated or a synergistic action, a significant beneficial effect on the cardiovascular system. With a significant increase in the generation of data on the dietary impact of flaxseed on the cardiovascular system, a review of where we stand - what we know and what we still need to understand about these effects on the heart and the vasculature - was thought to be of value and the rationale for this paper. For example, although we now know how to deliver the bioactives most efficiently (oil versus ground seed versus whole seed), we do not know how different foods can influence that delivery. Further, we know flaxseed has anti-arrhythmic, anti-atherogenic, anti-hypertensive, and cholesterol-lowering actions in animal studies and some selected human trials but much more needs to be learned, particularly in human trials. These results have justified further commitment of resources to the initiation of human trials. Because of the impact of nutrition on many chronic diseases, this may not only be true for the effects of flaxseed on cardiovascular disease but may be just as relevant for many other disease conditions.
Collapse
Affiliation(s)
- Mihir Parikh
- a Canadian Centre for Agri-food Research in Health and Medicine, Institute of Cardiovascular Sciences, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Colleges of Medicine and Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Grant N Pierce
- a Canadian Centre for Agri-food Research in Health and Medicine, Institute of Cardiovascular Sciences, St. Boniface Hospital, Winnipeg, MB R2H 2A6, Canada.,b Department of Physiology and Pathophysiology, Colleges of Medicine and Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| |
Collapse
|