1
|
Zeng H, Zhao W, Fang L, Pan H, Huang S, Zeng X. Effect of Stellate Ganglion Block on Dysphagia and Cognitive Impairment in Cerebral Small Vessel Disease: A Randomized Controlled Study. JOURNAL OF SPEECH, LANGUAGE, AND HEARING RESEARCH : JSLHR 2024; 67:3660-3672. [PMID: 39270205 DOI: 10.1044/2024_jslhr-24-00145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2024]
Abstract
BACKGROUNDS Cerebral small vessel disease (CSVD) can potentially and frequently lead to dysphagia and cognitive impairment. Stellate ganglion block (SGB) can alleviate the symptoms by regulating neural pathways and improving cerebral blood circulation. OBJECTIVES This study aimed to explore the clinical effect of SGB on airway protection, dysphagia, cognitive impairment, and activities of daily living (ADL) in CSVD patients. METHOD This was a randomized controlled study conducted from February 2021 to May 2023, including 84 CSVD patients with dysphagia and cognitive impairment. The participants were randomly divided into the SGB group (n = 42) and the untreated group (n = 42). All received standard-of-care rehabilitation for 20 days. The SGB group received SGB once a day additionally. Assessments were conducted on Day 1 and Day 20, respectively. The Penetration-Aspiration Scale (PAS) was primary outcome. Modified Barium Swallow Impairment (MBSImp), Mini-Mental State Examination (MMSE), Modified Barthel Index (MBI) and adverse events were secondary outcomes. This study was registered at ClinicalTrials.gov, Identifier: NCT06176404. RESULTS There were two dropout cases in the untreated group. Time effect with statistical significance was observed in all assessments (p < .05). Group effect with statistical significance was observed in the PAS (z = -17.283, p < .001), MBSImp-Oral (z = -3.382, p = .001), MBSImp-Pharyngeal (z = -2.639, p = .008), MMSE (F = 7.450, p = .008), and MBI (F = 6.408, p = .013). During the treatment, there were no severe adverse events. CONCLUSION SGB can significantly and safely improve airway protection, dysphagia, cognitive function, and ADL in CSVD patients with dysphagia and cognitive impairment who received standard-of-care rehabilitation.
Collapse
Affiliation(s)
- Hongji Zeng
- School of Public Health, Zhengzhou University, China
| | - Weijia Zhao
- School of Public Health, Zhengzhou University, China
| | - Lixuan Fang
- Department of Rehabilitation Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Huoying Pan
- First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | | | - Xi Zeng
- Department of Rehabilitation Medicine, The First Affiliated Hospital of Zhengzhou University, China
- NHC Key Laboratory of Prevention and Treatment of Cerebrovascular Diseases, Zhengzhou, China
| |
Collapse
|
2
|
Yang Y, Yao Z, Huo L. Causal effect of circulating cytokines on MRI markers of cerebral small vessel disease: A mendelian randomization study. Cytokine 2024; 182:156713. [PMID: 39079216 DOI: 10.1016/j.cyto.2024.156713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/05/2024] [Accepted: 07/22/2024] [Indexed: 08/25/2024]
Abstract
BACKGROUND Previous observational studies have reported the correlation between circulating inflammatory cytokines and cerebral small vessel disease (CSVD). However, the causality of this association is uncertain. This study used Mendelian randomization to investigate the causal effect of circulating inflammatory cytokines on neuroimaging changes in CSVD. METHODS This study utilized genetic variances of 41 inflammatory cytokines and 3 neuroimaging markers of CSVD from genome-wide association studies to assess the causal effects in a two-sample Mendelian randomization approach. Inverse variance weighted analysis was used as the main analytical method, and sensitivity analysis was used to further validate the robustness of the results. RESULTS Increased IL-18 was associated with increased white matter hyperintensity (WMH) and mean diffusivity (MD) (β = 0.034, 95 % CI 0.002, 0.065, P=0.038, β = 0.157, 95 % CI 0.015, 0.299, P=0.030). However, increased IL-18 was associated with decreased fractional anisotropy (FA) (β = -0.141, 95 % CI -0.279, -0.002, P=0.047). Increased monocyte chemotactic protein-1(MCP-1) was associated with decreased FA (β = -0.278, 95 % CI -0.502, -0.054, P=0.015). Increased IL-10 levels and IL-2ra levels were associated with decreased risks of MD (β = -0.228, 95 % CI -0.448, -0.009, p = 0.041; β = -0.204, 95 % CI=-0.377, -0.031, p = 0.021). CONCLUSIONS This study revealed that increased levels of IL-18 and MCP-1 were associated with white matter microstructural injury, and increased levels of IL-10 and IL-2ra were associated with decreased MD.
Collapse
Affiliation(s)
- Yang Yang
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Zhichao Yao
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China
| | - Lirong Huo
- Central Laboratory, Department of Neurology, Fu Xing Hospital, Capital Medical University, Beijing 100038, China.
| |
Collapse
|
3
|
Hou Y, Yan Z, Wan H, Yang J, Ding Z, He Y. A Combination of Astragaloside IV and Hydroxysafflor Yellow A Attenuates Cerebral Ischemia-Reperfusion Injury via NF-κB/NLRP3/Caspase-1/GSDMD Pathway. Brain Sci 2024; 14:781. [PMID: 39199474 PMCID: PMC11487458 DOI: 10.3390/brainsci14080781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
Cerebral ischemia-reperfusion injury (IRI), occurring after blood supply restoration, contributes significantly to stroke-related deaths. This study explored the combined impact and mechanisms of astragaloside IV (AS-IV), hydroxysafflor yellow A (HSYA), and their combination in mitigating IRI. Male Sprague-Dawley (SD) rats were randomized to the Sham, MCAO, MCAO+AS-IV, MCAO+HSYA, and MCAO+AS-IV+HSYA groups. Neurological deficits and cerebral infarction were examined after restoring the blood supply to the brain. Pathomorphological changes in the cerebral cortex were observed via HE staining. IL-1β and IL-18 were quantified using ELISA. The expression of NF-κB and GSDMD in the ischemic cerebrum was analyzed using immunohistochemistry. The expression levels of NLRP3, ASC, IL-1β, Caspase-1, and GSDMD in the ischemic cerebrum were evaluated using Western blot. The MCAO+AS-IV, MCAO+HSYA, and MCAO+AS-IV+HSYA groups exhibited notably better neurological function and cerebral infarction compared with the MCAO group. The combined treatment demonstrated superior brain tissue injury alleviation. Reductions in NF-κB, GSDMD positive cells, and NLRP3/ASC/IL-1β/Caspase-1/GSDMD protein expression in the ischemic brain were significantly more pronounced with the combined therapy, indicating a synergistic effect in countering cerebral IRI via the NF-κB/NLRP3/Caspase-1/GSDMD pathway inhibition of cell pyroptosis-induced injury.
Collapse
Affiliation(s)
- Yongchun Hou
- Basic Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou 310053, China
| | - Zi Yan
- Department of Basic Medicine, Nanchang Medical College, Nanchang 360000, China
| | - Haitong Wan
- Basic Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou 310053, China
| | - Jiehong Yang
- Basic Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou 310053, China
| | - Zhishan Ding
- Basic Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou 310053, China
| | - Yu He
- Basic Medical School, Zhejiang Chinese Medical University, Hangzhou 310053, China
- Key Laboratory of TCM Encephalopathy of Zhejiang Province, Hangzhou 310053, China
| |
Collapse
|
4
|
Duan Z, Jia W, Wang J, Xu D, Yang Y, Qi Z, Yang L, Wu C. Exploring the mechanism of Panax notoginseng saponin in inhibiting the inflammatory response of microglia in cerebral ischemia based on network pharmacology. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1566-1570. [PMID: 38973343 DOI: 10.3724/abbs.2024114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/09/2024] Open
Affiliation(s)
- Zhaoda Duan
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Wenji Jia
- Department of Neurology, No.2 Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Jianxiang Wang
- Ganmei Hospital Affiliated to Kunming Medical University, Kunming 650000, China
| | - Dongyao Xu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Yujia Yang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Zhi Qi
- Department of Neurology, No.2 Affiliated Hospital, Kunming Medical University, Kunming 650101, China
| | - Li Yang
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| | - Chunyun Wu
- Department of Anatomy and Histology/Embryology, Faculty of Basic Medical Sciences, Kunming Medical University, Kunming 650500, China
| |
Collapse
|
5
|
Jia R, Solé-Guardia G, Kiliaan AJ. Blood-brain barrier pathology in cerebral small vessel disease. Neural Regen Res 2024; 19:1233-1240. [PMID: 37905869 PMCID: PMC11467932 DOI: 10.4103/1673-5374.385864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/27/2023] [Accepted: 08/22/2023] [Indexed: 11/02/2023] Open
Abstract
ABSTRACT Cerebral small vessel disease is a neurological disease that affects the brain microvasculature and which is commonly observed among the elderly. Although at first it was considered innocuous, small vessel disease is nowadays regarded as one of the major vascular causes of dementia. Radiological signs of small vessel disease include small subcortical infarcts, white matter magnetic resonance imaging hyperintensities, lacunes, enlarged perivascular spaces, cerebral microbleeds, and brain atrophy; however, great heterogeneity in clinical symptoms is observed in small vessel disease patients. The pathophysiology of these lesions has been linked to multiple processes, such as hypoperfusion, defective cerebrovascular reactivity, and blood-brain barrier dysfunction. Notably, studies on small vessel disease suggest that blood-brain barrier dysfunction is among the earliest mechanisms in small vessel disease and might contribute to the development of the hallmarks of small vessel disease. Therefore, the purpose of this review is to provide a new foundation in the study of small vessel disease pathology. First, we discuss the main structural domains and functions of the blood-brain barrier. Secondly, we review the most recent evidence on blood-brain barrier dysfunction linked to small vessel disease. Finally, we conclude with a discussion on future perspectives and propose potential treatment targets and interventions.
Collapse
Affiliation(s)
- Ruxue Jia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Gemma Solé-Guardia
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| | - Amanda J. Kiliaan
- Department of Medical Imaging, Anatomy, Radboud University Medical Center, Donders Institute for Brain, Cognition & Behavior, Center for Medical Neuroscience, Preclinical Imaging Center PRIME, Radboud Alzheimer Center, Nijmegen, the Netherlands
| |
Collapse
|
6
|
Blaustein JR, Quisel MJ, Hamburg NM, Wittkopp S. Environmental Impacts on Cardiovascular Health and Biology: An Overview. Circ Res 2024; 134:1048-1060. [PMID: 38662864 PMCID: PMC11058466 DOI: 10.1161/circresaha.123.323613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
Environmental stressors associated with human activities (eg, air and noise pollution, light disturbance at night) and climate change (eg, heat, wildfires, extreme weather events) are increasingly recognized as contributing to cardiovascular morbidity and mortality. These harmful exposures have been shown to elicit changes in stress responses, circadian rhythms, immune cell activation, and oxidative stress, as well as traditional cardiovascular risk factors (eg, hypertension, diabetes, obesity) that promote cardiovascular diseases. In this overview, we summarize evidence from human and animal studies of the impacts of environmental exposures and climate change on cardiovascular health. In addition, we discuss strategies to reduce the impact of environmental risk factors on current and future cardiovascular disease burden, including urban planning, personal monitoring, and mitigation measures.
Collapse
Affiliation(s)
- Jacob R. Blaustein
- New York University Grossman School of Medicine, Department of Medicine, Leon H. Charney Division of Cardiology, New York, USA
| | - Matthew J. Quisel
- Department of Medicine, Boston University Chobanian and Avedision School of Medicine
| | - Naomi M. Hamburg
- Section of Vascular Biology, Whitaker Cardiovascular Institute, Chobanian and Avedisian School of Medicine, Boston University, Boston, USA
| | - Sharine Wittkopp
- New York University Grossman School of Medicine, Department of Medicine, Leon H. Charney Division of Cardiology, New York, USA
| |
Collapse
|
7
|
Gu J, Liu C, Yao Y. Prognostic potency of plasma LRG1 measurement at multiple time points in acute ischemic stroke patients. Biomark Med 2024; 18:181-190. [PMID: 38440887 DOI: 10.2217/bmm-2023-0545] [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] [Indexed: 03/06/2024] Open
Abstract
Objective: This study aimed to investigate the prognostic potency of LRG1 in acute ischemic stroke (AIS) patients. Methods: Plasma LRG1 levels were detected at admission and on days 3, 7 and 30 in 150 AIS patients. Results: LRG1 positively correlated with total cholesterol (p = 0.016), triglycerides (p = 0.046), C-reactive protein (p < 0.001), TNF-α (p = 0.001) and IL-6 (p = 0.004). After admission, LRG1 showed a decreasing trend (p < 0.001). Interestingly, LRG1 levels at admission (p = 0.014), day 3 (p = 0.027), day 7 (p = 0.008) and day 30 (p = 0.002) were higher in patients with modified Rankin scale score ≥2 versus those with scores <2. The LRG1 levels at day 7 (p = 0.032) and day 30 (p = 0.023) were higher in patients with recurrence versus no recurrence. Conclusion: LRG1 correlates with blood lipids, inflammation and short-term prognosis of AIS.
Collapse
Affiliation(s)
- Juxian Gu
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061001, China
| | - Chao Liu
- Department of CT Diagnosis, Cangzhou Central Hospital, Cangzhou, 061001, China
| | - Yan Yao
- Department of Neurology, Cangzhou Central Hospital, Cangzhou, 061001, China
| |
Collapse
|
8
|
Guo W, Huang D, Li S. Lycopene alleviates oxidative stress-induced cell injury in human vascular endothelial cells by encouraging the SIRT1/Nrf2/HO-1 pathway. Clin Exp Hypertens 2023; 45:2205051. [PMID: 37120838 DOI: 10.1080/10641963.2023.2205051] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
BACKGROUND AND OBJECTIVE Epidemiological research have displayed that dietary intake rich in lycopene, an antioxidant, is negatively correlated with the risk of cardiovascular disease (CVD). This study aimed to investigate whether the intervention with different concentrations of lycopene could attenuate H2O2-induced oxidative stress injury in human vascular endothelial cells (VECs). METHODS The human VECs HMEC-1 and ECV-304 were incubated with a final concentration of 300 µmol/L H2O2, followed by they were incubated with lycopene at doses of 0.5, 1, or 2 µm. Subsequently, cell proliferation, cytotoxicity, cell adhesion, reactive oxygen species (ROS) contents, adhesion molecule expression, oxidative stress levels, pro-inflammatory factor production, the apoptosis protein levels, and the silent information regulator-1 (SIRT1)/nuclear factor erythroid 2-related factor 2 (Nrf2)/heme oxygenase-1 (HO-1) pathway protein levels were tested by CCK-8 kit, lactate dehydrogenase (LDH) kit, immunofluorescence labeling, cell surface enzyme immunoassays (EIA), enzyme-linked immunosorbent assay (ELISA), as well as Western blot assays, respectively. RESULTS Under H2O2 stimulation, HMEC-1 and ECV-304 cell proliferation and the SIRT1/Nrf2/HO-1 pathway protein expression were significantly reduced, whereas cytotoxicity, apoptosis, cell adhesion molecule expression, pro-inflammatory and oxidative stress factors production were apparently encouraged, which were partially countered by lycopene intervention in a dose-dependent manner. CONCLUSION Lycopene alleviates H2O2-induced oxidative damage in human VECs by reducing intracellular ROS levels, inflammatory factor production, cell adhesiveness, and apoptosis rate under oxidative stress conditions through activation of the SIRT1/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Wenhai Guo
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Danping Huang
- The First Clinical Medicine School, Guangdong Pharmaceutical University, Guangzhou, 510006, China
| | - Shaodong Li
- Department of Traditional Chinese Medicine, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510000, China
- First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| |
Collapse
|
9
|
Hood RJ, Sanchez-Bezanilla S, Beard DJ, Rust R, Turner RJ, Stuckey SM, Collins-Praino LE, Walker FR, Nilsson M, Ong LK. Leakage beyond the primary lesion: A temporal analysis of cerebrovascular dysregulation at sites of hippocampal secondary neurodegeneration following cortical photothrombotic stroke. J Neurochem 2023; 167:733-752. [PMID: 38010732 DOI: 10.1111/jnc.16008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Revised: 10/10/2023] [Accepted: 10/11/2023] [Indexed: 11/29/2023]
Abstract
We have previously demonstrated that a cortical stroke causes persistent impairment of hippocampal-dependent cognitive tasks concomitant with secondary neurodegenerative processes such as amyloid-β accumulation in the hippocampus, a region remote from the primary infarct. Interestingly, there is emerging evidence suggesting that deposition of amyloid-β around cerebral vessels may lead to cerebrovascular structural changes, neurovascular dysfunction, and disruption of blood-brain barrier integrity. However, there is limited knowledge about the temporal changes of hippocampal cerebrovasculature after cortical stroke. In the current study, we aimed to characterise the spatiotemporal cerebrovascular changes after cortical stroke. This was done using the photothrombotic stroke model targeting the motor and somatosensory cortices of mice. Cerebrovascular morphology as well as the co-localisation of amyloid-β with vasculature and blood-brain barrier integrity were assessed in the cortex and hippocampal regions at 7, 28 and 84 days post-stroke. Our findings showed transient cerebrovascular remodelling in the peri-infarct area up to 28 days post-stroke. Importantly, the cerebrovascular changes were extended beyond the peri-infarct region to the ipsilateral hippocampus and were sustained out to 84 days post-stroke. When investigating vessel diameter, we showed a decrease at 84 days in the peri-infarct and CA1 regions that were exacerbated in vessels with amyloid-β deposition. Lastly, we showed sustained vascular leakage in the peri-infarct and ipsilateral hippocampus, indicative of a compromised blood-brain-barrier. Our findings indicate that hippocampal vasculature may represent an important therapeutic target to mitigate the progression of post-stroke cognitive impairment.
Collapse
Affiliation(s)
- Rebecca J Hood
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Sonia Sanchez-Bezanilla
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Daniel J Beard
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
| | - Ruslan Rust
- Institute for Regenerative Medicine (IREM), University of Zurich, Schlieren, Switzerland
- Department of Physiology and Neuroscience, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Renée J Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Shannon M Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Lyndsey E Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Frederick R Walker
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Centre for Rehab Innovations, The University of Newcastle, Callaghan, New South Wales, Australia
| | - Michael Nilsson
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- Centre for Rehab Innovations, The University of Newcastle, Callaghan, New South Wales, Australia
- School of Medicine and Public Health, The University of Newcastle, Callaghan, New South Wales, Australia
- LKC School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Lin Kooi Ong
- School of Biomedical Sciences and Pharmacy, The University of Newcastle, Callaghan, New South Wales, Australia
- Heart and Stroke Research Program, Hunter Medical Research Institute, New Lambton Heights, New South Wales, Australia
- School of Pharmacy, Monash University Malaysia, Bandar Sunway, Selangor, Malaysia
- School of Health and Medical Sciences & Centre for Health Research, University of Southern Queensland, Toowoomba, Queensland, Australia
| |
Collapse
|
10
|
Bains A, Sridhar K, Singh BN, Kuhad RC, Chawla P, Sharma M. Valorization of onion peel waste: From trash to treasure. CHEMOSPHERE 2023; 343:140178. [PMID: 37714483 DOI: 10.1016/j.chemosphere.2023.140178] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/22/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
Globally, fruits and vegetables are consumed as raw, processed, or as an additive, accounting for approximately 50% of total food wastage. Among the fruits and vegetables, onion is well known for its potential bioactive components; however, peels of onion are a major concern for the environmental health and food industries. Effective utilization methods for valorizing the onion peel should be needed to develop value-added products, which are more eco-friendly, cost-effective, and sustainable. Therefore, this review attempts to emphasize the conventional and emerging valorization techniques for onion peel waste to generate value-added products. Several vital applications including anticancerous, antiobesity, antimicrobial, and anti-inflammatory activities are thoroughly discussed. The findings showed that the use of advanced technologies like ultrasound-assisted extraction, microwave-assisted extraction, and enzymatic extraction, demonstrated improved extraction efficiency and higher yield of bioactive compounds, which showed the anticancerous, antiobesity, antimicrobial, and anti-inflammatory properties. However, in-depth studies are recommended to elucidate the mechanisms of action and potential synergistic effects of the bioactive compounds derived from onion peel waste, and to promote the sustainable utilization of onion peel waste in the long-term.
Collapse
Affiliation(s)
- Aarti Bains
- Department of Microbiology, Lovely Professional University, Phagwara, 144411, Punjab, India
| | - Kandi Sridhar
- Department of Food Technology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore, 641021, India
| | - Brahma Nand Singh
- Pharmacology Division, CSIR-National Botanical Research Institute, Lucknow, 226001, Uttar Pradesh, India
| | - Ramesh Chander Kuhad
- Sharda School of Basic Sciences and Research, Sharda University, Greater Noida - 201310, Uttar Pradesh, India; DPG Institute of Management and Technology, Sector-34, Gurugram - 122004, Haryana, India
| | - Prince Chawla
- Department of Food Technology and Nutrition, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Minaxi Sharma
- CARAH ASBL, Rue Paul Pastur, 11, Ath, 7800, Belgium.
| |
Collapse
|
11
|
Li W, Shao C, Li C, Zhou H, Yu L, Yang J, Wan H, He Y. Metabolomics: A useful tool for ischemic stroke research. J Pharm Anal 2023; 13:968-983. [PMID: 37842657 PMCID: PMC10568109 DOI: 10.1016/j.jpha.2023.05.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 05/14/2023] [Accepted: 05/29/2023] [Indexed: 10/17/2023] Open
Abstract
Ischemic stroke (IS) is a multifactorial and heterogeneous disease. Despite years of studies, effective strategies for the diagnosis, management and treatment of stroke are still lacking in clinical practice. Metabolomics is a growing field in systems biology. It is starting to show promise in the identification of biomarkers and in the use of pharmacometabolomics to help patients with certain disorders choose their course of treatment. The development of metabolomics has enabled further and more biological applications. Particularly, metabolomics is increasingly being used to diagnose diseases, discover new drug targets, elucidate mechanisms, and monitor therapeutic outcomes and its potential effect on precision medicine. In this review, we reviewed some recent advances in the study of metabolomics as well as how metabolomics might be used to identify novel biomarkers and understand the mechanisms of IS. Then, the use of metabolomics approaches to investigate the molecular processes and active ingredients of Chinese herbal formulations with anti-IS capabilities is summarized. We finally summarized recent developments in single cell metabolomics for exploring the metabolic profiles of single cells. Although the field is relatively young, the development of single cell metabolomics promises to provide a powerful tool for unraveling the pathogenesis of IS.
Collapse
Affiliation(s)
- Wentao Li
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chongyu Shao
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Chang Li
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Huifen Zhou
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Li Yu
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Jiehong Yang
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Haitong Wan
- School of Basic Medicine Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yu He
- School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| |
Collapse
|
12
|
Chuang SY, Hsu YC, Chou KW, Chang KS, Wong CH, Hsu YH, Cheng HM, Chen CW, Chen PY. Neutrophil-Lymphocyte Ratio as a Predictor of Cerebral Small Vessel Disease in a Geriatric Community: The I-Lan Longitudinal Aging Study. Brain Sci 2023; 13:1087. [PMID: 37509017 PMCID: PMC10377025 DOI: 10.3390/brainsci13071087] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/09/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cerebral Small Vessel Disease (CSVD) frequently affects the elderly, with inflammation playing a crucial role in related health complications, including dementia, stroke, and SVD. Studies, including animal experiments, indicate a strong link between inflammation and SVD progression. The Neutrophil-Lymphocyte Ratio (NLR) serves as a possible biomarker for ongoing inflammatory risks. A total of 720 adults aged 50 years or older from the community-based I-Lan Longitudinal Aging Study were included in this study. General linear regression and ordinally logistic regression analyses were performed to evaluate the association between NLR and CSVD. We further examined the presence of lacune, microbleed, and white matter hyperintensity (WMH) on brain MRI, which were used to construct a combined CSVD score. The NLR was positively associated with WMH (adjusted r = 0.109, p = 0.003), microbleed (adjusted r = 0.102, p = 0.006), and lacune (adjusted r = 0.100, p = 0.008). After adjustments for smoking, drinking, and physical activity in the ordinal logistic regression analysis, age, gender, brachial Systolic Blood Pressure (SBP), fasting glucose, LDL-cholesterol, and Hs-CRP were compared among subjects with low tertile (T1), medium tertile (T2) and high tertile (T3) NLR. The results showed that T2 vs. T1 had an odds ratio of 1.23 (0.86-1.77); and T3 vs. T1 had an odds ratio of 1.87 (1.29-2.71) of CSVD scores in four groups (zero (reference group), one, two, and three or more). NLR could be used to assess the state of inflammation in cerebral vessels. A significant and positive correlation between NLR and CSVD was verified in this study. However, the practical clinical application of NLR in CSVD patients and prognosis prediction should be validated through more scientific attempts.
Collapse
Affiliation(s)
- Shao-Yuan Chuang
- Institute of Population Health Science, National Health Research Institute, Miaoli 36001, Taiwan
- Institute of Public Health, National Yang Ming Chiao Tung University School of Medicine, Taipei 30010, Taiwan
| | - Yin-Chen Hsu
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital Chiayi Branch, Chiayi 61363, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Kuang-Wei Chou
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Department of Emergency Medicine, Mackay Memorial Hospital, Taipei 11008, Taiwan
| | - Kuo-Song Chang
- Department of Emergency Medicine, Mackay Memorial Hospital, Taipei 11008, Taiwan
- Mackay Junior College of Medicine, Nursing, and Management, Taipei 11260, Taiwan
| | - Chiong-Hee Wong
- Department of Emergency Medicine, Mackay Memorial Hospital, Taipei 11008, Taiwan
| | - Ya-Hui Hsu
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Department of Emergency Medicine, Mackay Memorial Hospital, Taipei 11008, Taiwan
| | - Hao-Min Cheng
- Institute of Public Health, National Yang Ming Chiao Tung University School of Medicine, Taipei 30010, Taiwan
- Department of Internal Medicine, National Yang Ming Chiao Tung University School of Medicine, Taipei 31254, Taiwan
- Center for Evidence-Based Medicine, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Medical Education, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chien-Wei Chen
- Department of Diagnostic Radiology, Chang Gung Memorial Hospital Chiayi Branch, Chiayi 61363, Taiwan
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Pang-Yen Chen
- Institute of Public Health, National Yang Ming Chiao Tung University School of Medicine, Taipei 30010, Taiwan
- Department of Nursing, Yuanpei University of Medical Technology, Hsinchu 30015, Taiwan
- Department of Emergency Medicine, Mackay Memorial Hospital, Taipei 11008, Taiwan
| |
Collapse
|
13
|
Liu W, Wang X, Feng Y. Restoring endothelial function: shedding light on cardiovascular stent development. Biomater Sci 2023. [PMID: 37161519 DOI: 10.1039/d3bm00390f] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
Complete endothelialization is highly important for maintaining long-term patency and avoiding subsequent complications in implanting cardiovascular stents. It not only refers to endothelial cells (ECs) fully covering the inserted stents, but also includes the newly formed endothelium, which could exert physiological functions, such as anti-thrombosis and anti-stenosis. Clinical outcomes have indicated that endothelial dysfunction, especially the insufficiency of antithrombotic and barrier functions, is responsible for stent failure. Learning from vascular pathophysiology, endothelial dysfunction on stents is closely linked to the microenvironment of ECs. Evidence points to inflammatory responses, oxidative stress, altered hemodynamic shear stress, and impaired endothelial barrier affecting the normal growth of ECs, which are the four major causes of endothelial dysfunction. The related molecular mechanisms and efforts dedicated to improving the endothelial function are emphasized in this review. From the perspective of endothelial function, the design principles, advantages, and disadvantages behind current stents are introduced to enlighten the development of new-generation stents, aiming to offer new alternatives for restoring endothelial function.
Collapse
Affiliation(s)
- Wen Liu
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, P. R. China
| | - Xiaoyu Wang
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, P. R. China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, P. R. China.
- Collaborative Innovation Center of Chemical Science and Chemical Engineering (Tianjin), Weijin Road 92, Tianjin 300072, P. R. China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Weijin Road 92, Tianjin 300072, P. R. China
- Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin 300072, China
| |
Collapse
|
14
|
Astigiano C, Piacente F, Laugieri ME, Benzi A, Di Buduo CA, Miguel CP, Soncini D, Cea M, Antonelli A, Magnani M, Balduini A, De Flora A, Bruzzone S. Sirtuin 6 Regulates the Activation of the ATP/Purinergic Axis in Endothelial Cells. Int J Mol Sci 2023; 24:ijms24076759. [PMID: 37047732 PMCID: PMC10095398 DOI: 10.3390/ijms24076759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/03/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
Sirtuin 6 (SIRT6) is a member of the mammalian NAD+-dependent deac(et)ylase sirtuin family. SIRT6’s anti-inflammatory roles are emerging increasingly often in different diseases and cell types, including endothelial cells. In this study, the role of SIRT6 in pro-inflammatory conditions was investigated by engineering human umbilical vein endothelial cells to overexpress SIRT6 (SIRT6+ HUVECs). Our results showed that SIRT6 overexpression affected the levels of adhesion molecules and sustained megakaryocyte proliferation and proplatelet formation. Interestingly, the pro-inflammatory activation of the ATP/purinergic axis was reduced in SIRT6+ HUVECs. Specifically, the TNFα-induced release of ATP in the extracellular space and the increase in pannexin-1 hemichannel expression, which mediates ATP efflux, were hampered in SIRT6+ cells. Instead, NAD+ release and Connexin43 expression were not modified by SIRT6 levels. Moreover, the Ca2+ influx in response to ATP and the expression of the purinergic receptor P2X7 were decreased in SIRT6+ HUVECs. Contrary to extracellular ATP, extracellular NAD+ did not evoke pro-inflammatory responses in HUVECs. Instead, NAD+ administration reduced endothelial cell proliferation and motility and counteracted the TNFα-induced angiogenesis. Altogether, our data reinforce the view of SIRT6 activation as an anti-inflammatory approach in vascular endothelium.
Collapse
Affiliation(s)
- Cecilia Astigiano
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Francesco Piacente
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Maria Elena Laugieri
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Andrea Benzi
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Christian A. Di Buduo
- Department of Molecular Medicine, University of Pavia, Via C. Forlanini 6, 27100 Pavia, Italy
| | - Carolina P. Miguel
- Department of Molecular Medicine, University of Pavia, Via C. Forlanini 6, 27100 Pavia, Italy
| | - Debora Soncini
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
| | - Michele Cea
- Department of Internal Medicine, University of Genova, Viale Benedetto XV, 6, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 16132 Genova, Italy
| | - Antonella Antonelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Via Saffi 2, 61029 Urbino, Italy
| | - Alessandra Balduini
- Department of Molecular Medicine, University of Pavia, Via C. Forlanini 6, 27100 Pavia, Italy
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, MA 02155, USA
| | - Antonio De Flora
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
| | - Santina Bruzzone
- Department of Experimental Medicine, University of Genova, Viale Benedetto XV, 1, 16132 Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Largo R. Benzi, 16132 Genova, Italy
| |
Collapse
|
15
|
Burda R, Burda J, Morochovič R. Ischemic Tolerance—A Way to Reduce the Extent of Ischemia–Reperfusion Damage. Cells 2023; 12:cells12060884. [PMID: 36980225 PMCID: PMC10047660 DOI: 10.3390/cells12060884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/08/2023] [Accepted: 03/11/2023] [Indexed: 03/14/2023] Open
Abstract
Individual tissues have significantly different resistance to ischemia–reperfusion damage. There is still no adequate treatment for the consequences of ischemia–reperfusion damage. By utilizing ischemic tolerance, it is possible to achieve a significant reduction in the extent of the cell damage due to ischemia–reperfusion injury. Since ischemia–reperfusion damage usually occurs unexpectedly, the use of preconditioning is extremely limited. In contrast, postconditioning has wider possibilities for use in practice. In both cases, the activation of ischemic tolerance can also be achieved by the application of sublethal stress on a remote organ. Despite very encouraging and successful results in animal experiments, the clinical results have been disappointing so far. To avoid the factors that prevent the activation of ischemic tolerance, the solution has been to use blood plasma containing tolerance effectors. This plasma is taken from healthy donors in which, after exposure to two sublethal stresses within 48 h, effectors of ischemic tolerance occur in the plasma. Application of this activated plasma to recipient animals after the end of lethal ischemia prevents cell death and significantly reduces the consequences of ischemia–reperfusion damage. Until there is a clear chemical identification of the end products of ischemic tolerance, the simplest way of enhancing ischemic tolerance will be the preparation of activated plasma from young healthy donors with the possibility of its immediate use in recipients during the initial treatment.
Collapse
Affiliation(s)
- Rastislav Burda
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
- Correspondence:
| | - Jozef Burda
- Institute of Neurobiology, Slovak Academy of Sciences, 040 01 Košice, Slovakia
| | - Radoslav Morochovič
- Department of Trauma Surgery, Faculty of Medicine, Pavol Jozef Šafárik University in Košice, Rastislavova 43, 040 01 Košice, Slovakia
- Department of Trauma Surgery, Louis Pasteur University Hospital, Rastislavova 43, 040 01 Košice, Slovakia
| |
Collapse
|
16
|
Huang Y, Omorou M, Gao M, Mu C, Xu W, Xu H. Hydrogen sulfide and its donors for the treatment of cerebral ischaemia-reperfusion injury: A comprehensive review. Biomed Pharmacother 2023; 161:114506. [PMID: 36906977 DOI: 10.1016/j.biopha.2023.114506] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/12/2023] Open
Abstract
As an endogenous gas signalling molecule, hydrogen sulfide (H2S) is frequently present in a variety of mammals and plays a significant role in the cardiovascular and nervous systems. Reactive oxygen species (ROS) are produced in large quantities as a result of cerebral ischaemia-reperfusion, which is a very serious class of cerebrovascular diseases. ROS cause oxidative stress and induce specific gene expression that results in apoptosis. H2S reduces cerebral ischaemia-reperfusion-induced secondary injury via anti-oxidative stress injury, suppression of the inflammatory response, inhibition of apoptosis, attenuation of cerebrovascular endothelial cell injury, modulation of autophagy, and antagonism of P2X7 receptors, and it plays an important biological role in other cerebral ischaemic injury events. Despite the many limitations of the hydrogen sulfide therapy delivery strategy and the difficulty in controlling the ideal concentration, relevant experimental evidence demonstrating that H2S plays an excellent neuroprotective role in cerebral ischaemia-reperfusion injury (CIRI). This paper examines the synthesis and metabolism of the gas molecule H2S in the brain as well as the molecular mechanisms of H2S donors in cerebral ischaemia-reperfusion injury and possibly other unknown biological functions. With the active development in this field, it is expected that this review will assist researchers in their search for the potential value of hydrogen sulfide and provide new ideas for preclinical trials of exogenous H2S.
Collapse
Affiliation(s)
- Yiwei Huang
- Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China; Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| | - Moussa Omorou
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Meng Gao
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Chenxi Mu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China; Basic Medical College, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Weijing Xu
- School of Public Health, Jiamusi University, Jiamusi 154007, Heilongjiang, China.
| | - Hui Xu
- Key Laboratory of Microecology-Immune Regulatory Network and Related Diseases, Jiamusi 154007, Heilongjiang, China.
| |
Collapse
|
17
|
Purinergic receptors mediate endothelial dysfunction and participate in atherosclerosis. Purinergic Signal 2023; 19:265-272. [PMID: 34981330 PMCID: PMC9984579 DOI: 10.1007/s11302-021-09839-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 12/20/2021] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis is the main pathological basis of cardiovascular disease and involves damage to vascular endothelial cells (ECs) that results in endothelial dysfunction (ED). The vascular endothelium is the key to maintaining blood vessel health and homeostasis. ED is a complex pathological process involving inflammation, shear stress, vascular tone, adhesion of leukocytes to ECs, and platelet aggregation. The activation of P2X4, P2X7, and P2Y2 receptors regulates vascular tone in response to shear stress, while activation of the A2A, P2X4, P2X7, P2Y1, P2Y2, P2Y6, and P2Y12 receptors promotes the secretion of inflammatory cytokines. Finally, P2X1, P2Y1, and P2Y12 receptor activation regulates platelet activity. These purinergic receptors mediate ED and participate in atherosclerosis. In short, P2X4, P2X7, P2Y1, and P2Y12 receptors are potential therapeutic targets for atherosclerosis.
Collapse
|
18
|
Wang Y, Zhu Y, Wang J, Dong L, Liu S, Li S, Wu Q. Purinergic signaling: A gatekeeper of blood-brain barrier permeation. Front Pharmacol 2023; 14:1112758. [PMID: 36825149 PMCID: PMC9941648 DOI: 10.3389/fphar.2023.1112758] [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: 11/30/2022] [Accepted: 01/27/2023] [Indexed: 02/10/2023] Open
Abstract
This review outlined evidence that purinergic signaling is involved in the modulation of blood-brain barrier (BBB) permeability. The functional and structural integrity of the BBB is critical for maintaining the homeostasis of the brain microenvironment. BBB integrity is maintained primarily by endothelial cells and basement membrane but also be regulated by pericytes, neurons, astrocytes, microglia and oligodendrocytes. In this review, we summarized the purinergic receptors and nucleotidases expressed on BBB cells and focused on the regulation of BBB permeability by purinergic signaling. The permeability of BBB is regulated by a series of purinergic receptors classified as P2Y1, P2Y4, P2Y12, P2X4, P2X7, A1, A2A, A2B, and A3, which serve as targets for endogenous ATP, ADP, or adenosine. P2Y1 and P2Y4 antagonists could attenuate BBB damage. In contrast, P2Y12-mediated chemotaxis of microglial cell processes is necessary for rapid closure of the BBB after BBB breakdown. Antagonists of P2X4 and P2X7 inhibit the activation of these receptors, reduce the release of interleukin-1 beta (IL-1β), and promote the function of BBB closure. In addition, the CD39/CD73 nucleotidase axis participates in extracellular adenosine metabolism and promotes BBB permeability through A1 and A2A on BBB cells. Furthermore, A2B and A3 receptor agonists protect BBB integrity. Thus, the regulation of the BBB by purinergic signaling is complex and affects the opening and closing of the BBB through different pathways. Appropriate selective agonists/antagonists of purinergic receptors and corresponding enzyme inhibitors could modulate the permeability of the BBB, effectively delivering therapeutic drugs/cells to the central nervous system (CNS) or limiting the entry of inflammatory immune cells into the brain and re-establishing CNS homeostasis.
Collapse
Affiliation(s)
| | | | - Junmeng Wang
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Longcong Dong
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuqing Liu
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Sihui Li
- Acupuncture and Moxibustion College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | | |
Collapse
|
19
|
The NLRP3 Inflammasome in Age-Related Cerebral Small Vessel Disease Manifestations: Untying the Innate Immune Response Connection. LIFE (BASEL, SWITZERLAND) 2023; 13:life13010216. [PMID: 36676165 PMCID: PMC9866483 DOI: 10.3390/life13010216] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/07/2023] [Accepted: 01/09/2023] [Indexed: 01/14/2023]
Abstract
In this narrative review, we present the evidence on nucleotide-binding and oligomerization (NOD) domain-like receptor (NLR) family pyrin domain (PYD)-containing 3 (NLRP3) inflammasome activation for its putative roles in the elusive pathomechanism of aging-related cerebral small vessel disease (CSVD). Although NLRP3 inflammasome-interleukin (IL)-1β has been implicated in the pathophysiology of coronary artery disease, its roles in cerebral arteriothrombotic micro-circulation disease such as CSVD remains unexplored. Here, we elaborate on the current manifestations of CSVD and its' complex pathogenesis and relate the array of activators and aberrant activation involving NLRP3 inflammasome with this condition. These neuroinflammatory insights would expand on our current understanding of CSVD clinical (and subclinical) heterogenous manifestations whilst highlighting plausible NLRP3-linked therapeutic targets.
Collapse
|
20
|
Kubatka P, Mazurakova A, Koklesova L, Samec M, Sokol J, Samuel SM, Kudela E, Biringer K, Bugos O, Pec M, Link B, Adamkov M, Smejkal K, Büsselberg D, Golubnitschaja O. Antithrombotic and antiplatelet effects of plant-derived compounds: a great utility potential for primary, secondary, and tertiary care in the framework of 3P medicine. EPMA J 2022; 13:407-431. [PMID: 35990779 PMCID: PMC9376584 DOI: 10.1007/s13167-022-00293-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 12/29/2022]
Abstract
Thromboembolism is the third leading vascular disease, with a high annual incidence of 1 to 2 cases per 1000 individuals within the general population. The broader term venous thromboembolism generally refers to deep vein thrombosis, pulmonary embolism, and/or a combination of both. Therefore, thromboembolism can affect both - the central and peripheral veins. Arterial thromboembolism causes systemic ischemia by disturbing blood flow and oxygen supply to organs, tissues, and cells causing, therefore, apoptosis and/or necrosis in the affected tissues. Currently applied antithrombotic drugs used, e.g. to protect affected individuals against ischemic stroke, demonstrate significant limitations. For example, platelet inhibitors possess only moderate efficacy. On the other hand, thrombolytics and anticoagulants significantly increase hemorrhage. Contextually, new approaches are extensively under consideration to develop next-generation antithrombotics with improved efficacy and more personalized and targeted application. To this end, phytochemicals show potent antithrombotic efficacy demonstrated in numerous in vitro, ex vivo, and in vivo models as well as in clinical evaluations conducted on healthy individuals and persons at high risk of thrombotic events, such as pregnant women (primary care), cancer, and COVID-19-affected patients (secondary and tertiary care). Here, we hypothesized that specific antithrombotic and antiplatelet effects of plant-derived compounds might be of great clinical utility in primary, secondary, and tertiary care. To increase the efficacy, precise patient stratification based on predictive diagnostics is essential for targeted protection and treatments tailored to the person in the framework of 3P medicine. Contextually, this paper aims at critical review toward the involvement of specific classes of phytochemicals in antiplatelet and anticoagulation adapted to clinical needs. The paper exemplifies selected plant-derived drugs, plant extracts, and whole plant foods/herbs demonstrating their specific antithrombotic, antiplatelet, and fibrinolytic activities relevant for primary, secondary, and tertiary care. One of the examples considered is antithrombotic and antiplatelet protection specifically relevant for COVID-19-affected patient groups.
Collapse
Affiliation(s)
- Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Alena Mazurakova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Lenka Koklesova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Marek Samec
- Department of Pathological Physiology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Juraj Sokol
- Department of Hematology and Transfusion Medicine, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, 24144 Doha, Qatar
| | - Erik Kudela
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Kamil Biringer
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | | | - Martin Pec
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Barbara Link
- Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| | - Marian Adamkov
- Department of Histology and Embryology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia
| | - Karel Smejkal
- Department of Natural Drugs, Faculty of Pharmacy, Masaryk University, 61200 Brno, Czech Republic
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Qatar Foundation, Education City, 24144 Doha, Qatar
| | - Olga Golubnitschaja
- Predictive, Preventive and Personalised (3P) Medicine, Department of Radiation Oncology, University Hospital Bonn, Rheinische Friedrich-Wilhelms-Universität Bonn, 53127 Bonn, Germany
| |
Collapse
|
21
|
Wittkopp S, Wu F, Windheim J, Robinson M, Kannan K, Katz SD, Chen Y, Newman JD. Vascular endothelium as a target for perfluroalkyl substances (PFAs). ENVIRONMENTAL RESEARCH 2022; 212:113339. [PMID: 35447152 PMCID: PMC9233033 DOI: 10.1016/j.envres.2022.113339] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 04/15/2022] [Accepted: 04/17/2022] [Indexed: 06/14/2023]
Abstract
INTRODUCTION Perfluoroalkyl substances (PFAs) are ubiquitous, anthropogenic organic compounds that have been linked with cardiovascular disease and cardiovascular risk factors. Older, long-chain PFAs have been phased out due to adverse cardiometabolic health effect and replaced by newer short-chain PFAs. However, emerging research suggests that short-chain PFAs may also have adverse cardiovascular effects. Non-invasive measures of vascular function can detect preclinical cardiovascular disease and serve as a useful surrogate for early CVD risk. We hypothesized that serum concentrations of PFAs would be associated with noninvasive measures of vascular function, carotid-femoral pulse wave velocity (PWV) and brachial artery reactivity testing (BART), in adults with non-occupational exposure to PFAs. METHODS We measured serum concentrations of 14 PFAs with hybrid solid-phase extraction and ultrahigh-performance liquid chromatography-tandem mass spectrometry in 94 adult outpatients with no known cardiovascular disease. We collected clinical and demographic data; and measured vascular function, PWV and BART, using standard protocols. We assessed associations of individual PFAs with log-transformed BART and PWV using linear regression. We used weighted quantile sum regression to assess effects of correlated PFA mixtures on BART and PWV. RESULTS Ten PFAs were measured above the limit of detection in >50% of participants. Each standard deviation increase in concentration of perfluoroheptanoic acid (PFHpA) was associated with 15% decrease in BART (95% CI: -28.5, -0.17). The weighted index of a mixture of PFAs with correlated concentrations was inversely associated with BART: each tertile increase in the weighted PFA mixture was associated with 25% lower BART, with 73% of the effect driven by PFHpA. In contrast, no PFAs or mixtures were associated with PWV. CONCLUSIONS Serum concentration of PFHpA, a new, short-chain PFA, was associated with impaired vascular function among outpatients without CVD. Our findings support a potential adverse cardiovascular effect of newer, short-chain PFAs.
Collapse
Affiliation(s)
- Sharine Wittkopp
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY, 10016, USA
| | - Fen Wu
- Department of Population Health, New York University School of Medicine, 180 Madison Avenue, New York, NY, 10016, USA
| | - Joseph Windheim
- Department of Medicine, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Morgan Robinson
- Department of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA; Department of Pediatrics, New York University School of Medicine, 145 East 32 Street, 14th floor, New York, NY, 10016, USA
| | - Kurunthachalam Kannan
- Department of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA; Department of Pediatrics, New York University School of Medicine, 145 East 32 Street, 14th floor, New York, NY, 10016, USA
| | - Stuart D Katz
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY, 10016, USA
| | - Yu Chen
- Department of Population Health, New York University School of Medicine, 180 Madison Avenue, New York, NY, 10016, USA; Department of Environmental Medicine, New York University School of Medicine, 341 East 25th Street, New York, NY, 10010, USA
| | - Jonathan D Newman
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University School of Medicine, 550 First Avenue New York, NY, 10016, USA.
| |
Collapse
|
22
|
Hou L, Zhang S, Qi D, Jia T, Wang H, Zhang W, Wei S, Xue C, Wang P. Correlation between neutrophil/lymphocyte ratio and cognitive impairment in cerebral small vessel disease patients: A retrospective study. Front Neurol 2022; 13:925218. [PMID: 35989913 PMCID: PMC9391025 DOI: 10.3389/fneur.2022.925218] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 07/12/2022] [Indexed: 11/13/2022] Open
Abstract
Background and objective The blood neutrophil/lymphocyte ratio (NLR) is an objective and convenient parameter of systemic inflammation. Elevated NLR is associated with an increased risk of mild cognitive impairment (CI) in the elderly. However, few data are available on the impact of the NLR on CI in patients with cerebral small vessel disease (CSVD). Methods A total of 66 CSVD subjects with CI and 81 CSVD subjects without CI were evaluated in this study. Clinical, laboratory, radiological, and cognitive parameters were collected. The NLR was obtained with the absolute neutrophil count being divided by the absolute lymphocyte count in fasting blood samples. Logistic regression analysis was performed to evaluate the factors associated with CI. Receiver operating characteristic curves were illustrated to predict factors associated with CI in patients with CSVD. Results The NLR of the CI group was significantly higher than that of subjects without CI (2.59 vs. 2.21, P = 0.003). In multivariate analysis, NLR was positively correlated to the CI (OR: 1.43, 95% CI: 1.05–1.96, P = 0.024). It was suggested that the optimum NLR cutoff point for CI was 1.89 with 69.7% sensitivity and 59.3% specificity. Subjects with NLR ≥ 1.89 showed higher possibilities of CI compared to those with NLR < 1.89 (OR: 3.38, 95% CI: 1.62–7.07). Conclusions Correlations were found between NLR and CI. Patients with CSVD who have higher NLR might have an increased risk of CI.
Collapse
Affiliation(s)
- Lan Hou
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
- Baoding City Key Laboratory of Neurological Diseases, Baoding, China
| | - Shuhan Zhang
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
| | - Dandan Qi
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
| | - Tongle Jia
- Baoding City Key Laboratory of Neurological Diseases, Baoding, China
- Department of Neurosurgery, Baoding No.1 Central Hospital, Baoding, China
| | - Huan Wang
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
| | - Wei Zhang
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
| | - Shuyan Wei
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
| | - Conglong Xue
- Department of General Surgery, Baoding No.1 Central Hospital, Baoding, China
| | - Pei Wang
- Department of Neurology, Baoding No.1 Central Hospital, Baoding, China
- Baoding City Key Laboratory of Neurological Diseases, Baoding, China
- *Correspondence: Pei Wang
| |
Collapse
|
23
|
Tang X, Liu H, Xiao Y, Wu L, Shu P. Vitamin C Intake and Ischemic Stroke. Front Nutr 2022; 9:935991. [PMID: 35911106 PMCID: PMC9330473 DOI: 10.3389/fnut.2022.935991] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/22/2022] [Indexed: 12/31/2022] Open
Abstract
Vitamin C is an essential micronutrient with important antioxidant properties. Ischemic stroke is a major public health problem worldwide. Extensive evidence demonstrates that vitamin C has protective effects against cardiovascular disease, and there is a close relationship between vitamin C intake and ischemic stroke risk. Based on the evidence, we conducted this umbrella review to clarify the relationship between vitamin C intake and ischemic stroke risk from four perspectives: cellular mechanisms, animal experiments, clinical trials, and cohort studies.
Collapse
Affiliation(s)
- Xiaolong Tang
- Department of Internal Neurology, Beilun District People's Hospital, Ningbo, China
| | - Hanguang Liu
- Department of Internal Neurology, Beilun District People's Hospital, Ningbo, China
| | - Yuan Xiao
- Department of Internal Neurology, Beilun District People's Hospital, Ningbo, China
| | - Lei Wu
- Department of Painology, The No. 1 People's Hospital of Ningbo, Ningbo, China
- Lei Wu
| | - Peng Shu
- Department of Molecular Laboratory, Beilun District People's Hospital, Ningbo, China
- *Correspondence: Peng Shu
| |
Collapse
|
24
|
Rovati G, Contursi A, Bruno A, Tacconelli S, Ballerini P, Patrignani P. Antiplatelet Agents Affecting GPCR Signaling Implicated in Tumor Metastasis. Cells 2022; 11:725. [PMID: 35203374 PMCID: PMC8870128 DOI: 10.3390/cells11040725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 02/10/2022] [Accepted: 02/16/2022] [Indexed: 11/16/2022] Open
Abstract
Metastasis requires that cancer cells survive in the circulation, colonize distant organs, and grow. Despite platelets being central contributors to hemostasis, leukocyte trafficking during inflammation, and vessel stability maintenance, there is significant evidence to support their essential role in supporting metastasis through different mechanisms. In addition to their direct interaction with cancer cells, thus forming heteroaggregates such as leukocytes, platelets release molecules that are necessary to promote a disseminating phenotype in cancer cells via the induction of an epithelial-mesenchymal-like transition. Therefore, agents that affect platelet activation can potentially restrain these prometastatic mechanisms. Although the primary adhesion of platelets to cancer cells is mainly independent of G protein-mediated signaling, soluble mediators released from platelets, such as ADP, thromboxane (TX) A2, and prostaglandin (PG) E2, act through G protein-coupled receptors (GPCRs) to cause the activation of more additional platelets and drive metastatic signaling pathways in cancer cells. In this review, we examine the contribution of the GPCRs of platelets and cancer cells in the development of cancer metastasis. Finally, the possible use of agents affecting GPCR signaling pathways as antimetastatic agents is discussed.
Collapse
Affiliation(s)
- Gianenrico Rovati
- Department of Pharmaceutical Sciences, University of Milan, 20122 Milan, Italy;
| | - Annalisa Contursi
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Annalisa Bruno
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Stefania Tacconelli
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Patrizia Ballerini
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Innovative Technologies in Medicine and Dentistry, “G. d’Annunzio” University, 66100 Chieti, Italy
| | - Paola Patrignani
- Laboratory of Systems Pharmacology and Translational Therapies, Center for Advanced Studies and Technology (CAST), School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy; (A.C.); (A.B.); (S.T.); (P.B.)
- Department of Neuroscience, Imaging and Clinical Science, School of Medicine, “G. d’Annunzio” University, 66100 Chieti, Italy
| |
Collapse
|
25
|
Stuckey SM, Ong LK, Collins-Praino LE, Turner RJ. Neuroinflammation as a Key Driver of Secondary Neurodegeneration Following Stroke? Int J Mol Sci 2021; 22:ijms222313101. [PMID: 34884906 PMCID: PMC8658328 DOI: 10.3390/ijms222313101] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 11/25/2021] [Accepted: 12/01/2021] [Indexed: 01/13/2023] Open
Abstract
Ischaemic stroke involves the rapid onset of focal neurological dysfunction, most commonly due to an arterial blockage in a specific region of the brain. Stroke is a leading cause of death and common cause of disability, with over 17 million people worldwide suffering from a stroke each year. It is now well-documented that neuroinflammation and immune mediators play a key role in acute and long-term neuronal tissue damage and healing, not only in the infarct core but also in distal regions. Importantly, in these distal regions, termed sites of secondary neurodegeneration (SND), spikes in neuroinflammation may be seen sometime after the initial stroke onset, but prior to the presence of the neuronal tissue damage within these regions. However, it is key to acknowledge that, despite the mounting information describing neuroinflammation following ischaemic stroke, the exact mechanisms whereby inflammatory cells and their mediators drive stroke-induced neuroinflammation are still not fully understood. As a result, current anti-inflammatory treatments have failed to show efficacy in clinical trials. In this review we discuss the complexities of post-stroke neuroinflammation, specifically how it affects neuronal tissue and post-stroke outcome acutely, chronically, and in sites of SND. We then discuss current and previously assessed anti-inflammatory therapies, with a particular focus on how failed anti-inflammatories may be repurposed to target SND-associated neuroinflammation.
Collapse
Affiliation(s)
- Shannon M. Stuckey
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Lin Kooi Ong
- School of Pharmacy, Monash University Malaysia, Subang Jaya 47500, Malaysia;
- School of Biomedical Sciences and Pharmacy and the Priority Research Centre for Stroke and Brain Injury, The University of Newcastle, Callaghan 2308, Australia
| | - Lyndsey E. Collins-Praino
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
| | - Renée J. Turner
- Discipline of Anatomy and Pathology, School of Biomedicine, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide 5005, Australia; (S.M.S.); (L.E.C.-P.)
- Correspondence: ; Tel.: +61-8-8313-3114
| |
Collapse
|