1
|
Nguyen TD, Winek MA, Rao MK, Dhyani SP, Lee MY. Nuclear envelope components in vascular mechanotransduction: emerging roles in vascular health and disease. Nucleus 2025; 16:2453752. [PMID: 39827403 DOI: 10.1080/19491034.2025.2453752] [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: 10/08/2024] [Revised: 01/08/2025] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The vascular network, uniquely sensitive to mechanical changes, translates biophysical forces into biochemical signals for vessel function. This process relies on the cell's architectural integrity, enabling uniform responses to physical stimuli. Recently, the nuclear envelope (NE) has emerged as a key regulator of vascular cell function. Studies implicate nucleoskeletal elements (e.g. nuclear lamina) and the linker of nucleoskeleton and cytoskeleton (LINC) complex in force transmission, emphasizing nucleo-cytoskeletal communication in mechanotransduction. The nuclear pore complex (NPC) and its component proteins (i.e. nucleoporins) also play roles in cardiovascular disease (CVD) progression. We herein summarize evidence on the roles of nuclear lamina proteins, LINC complex members, and nucleoporins in endothelial and vascular cell mechanotransduction. Numerous studies attribute NE components in cytoskeletal-related cellular behaviors to insinuate dysregulation of nucleocytoskeletal feedback and nucleocytoplasmic transport as a mechanism of endothelial and vascular dysfunction, and hence implications for aging and vascular pathophysiology.
Collapse
Affiliation(s)
- Tung D Nguyen
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Michael A Winek
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Mihir K Rao
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Shaiva P Dhyani
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| | - Monica Y Lee
- Department of Physiology and Biophysics, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
- The Center for Cardiovascular Research, The University of Illinois at Chicago - College of Medicine, Chicago, IL, USA
| |
Collapse
|
2
|
Li K, Wang S, Li J, Wang L, Zhang Q, Hou L, Yu X, Liu Z, Lv T, Shang L. Low shear stress induces vascular endothelial cells apoptosis via miR-330 /SOD2 /HSP70 signaling pathway. Exp Cell Res 2025; 445:114410. [PMID: 39788367 DOI: 10.1016/j.yexcr.2025.114410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 11/30/2024] [Accepted: 01/06/2025] [Indexed: 01/12/2025]
Abstract
Atherosclerosis (AS) is a chronic disease initiated by vascular endothelial dysfunction, with low shear stress (SS) being a critical inducing factor in this dysfunction. Apoptosis, a form of programmed cell death, is closely associated with AS progression. However, the impact of low SS on endothelial apoptosis and its specific molecular mechanisms remains unclear. Our study revealed that low SS induces apoptosis in endothelial cells and contributes to endothelial dysfunction. Under low SS conditions, miR-330 expression was markedly upregulated, which subsequently targeted and inhibited SOD2 expression, leading to ROS accumulation and oxidative stress. Overexpression of SOD2 under low SS conditions markedly elevated HSP70 expression, contributing to endothelial homeostasis. However, when HSP70 expression was inhibited in the context of SOD2 overexpression, there was a significant increase in pro-apoptotic proteins (BAX and cleaved-caspase-3) and total apoptosis rate, along with a significant reduction in endothelial function markers such as nitric oxide and endothelial nitric oxide synthase. Notably, our experiments indicated no direct interaction between SOD2 and HSP70. Furthermore, inhibiting ROS production significantly raised HSP70 expression, suggesting that SOD2 regulates HSP70 in an indirect process involving ROS. In summary, our findings elucidate that low SS induces endothelial apoptosis and dysfunction through the miR-330/SOD2/HSP70 signaling pathway, providing valuable insights into AS intervention and prevention.
Collapse
Affiliation(s)
- Ke Li
- Department of Gastroenterology, 215 Hospital of Shaanxi Province, Xianyang, 712000, China; School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Shaohu Wang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Immunization and Planning, Heping District Center for Disease Control and Prevention, Tianjin, 300041, China
| | - Jiana Li
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Gastroenterology, No.983rd Hospital of the Chinese People's Liberation Army Joint Logistics and Security Forces, Tianjin, 300143, China
| | - Lingling Wang
- Department of Gastroenterology, 215 Hospital of Shaanxi Province, Xianyang, 712000, China
| | - Qin Zhang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Liming Hou
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Xinyi Yu
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Zhendong Liu
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China
| | - Ting Lv
- Department of Gastroenterology, 215 Hospital of Shaanxi Province, Xianyang, 712000, China.
| | - Luxiang Shang
- School of Clinical and Basic Medical Sciences, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, 250117, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, 250014, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250117, China.
| |
Collapse
|
3
|
Hoyek S, Yuan M, Bejjani R, Zacks DN, Miller JW, Vavvas DG, Patel NA. Multimodal Imaging Characteristics and Correlation to Outcomes in Patients With Central Retinal Artery Occlusion Presenting to a Large Academic Center. Am J Ophthalmol 2025; 270:35-51. [PMID: 39413899 DOI: 10.1016/j.ajo.2024.10.004] [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: 07/29/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/18/2024]
Abstract
PURPOSE To characterize a large modern cohort of patients with central retinal artery occlusion (CRAO) by describing presenting features and outcomes relating to manually segmented optical coherence tomography (OCT) features, angiographic reperfusion, and visual recovery. DESIGN Retrospective clinical cohort study. METHODS Patients with CRAO (ICD-10: H34.1) initially presenting to a tertiary referral center between January 2017 and December 2021 were included. Demographics, eye exam findings, fundus photographs, OCT, and fluorescein angiography were analyzed. Main outcome measures included total and inner retinal thickness on macular OCT, reperfusion, visual outcomes, and development of neovascularization. RESULTS A total of 145 eyes of 144 patients with mean age at of 69.4 ± 13.6 years were included. The mean time to presentation was 1.6 ± 4.2 days, with 19% examined within 4.5 hours and 26% within 6 hours of vision loss. 19% had cilioretinal artery (CLRA) sparing. Mean initial visual acuity (VA) was 1.68 ± 1.10 Logarithm of the Minimum Angle of Resolution (LogMAR) (CLRA sparing) compared to 2.53 ± 0.58 LogMAR (non-CLRA sparing), P < .001. 32% had elevated inflammatory makers. Out of 47 eyes with final fluorescein angiography, one-third showed some reperfusion. Final vision was 1.40 ± 1.16 LogMAR (CLRA sparing) compared to 2.46 ± 0.81 (non-CLRA sparing), P < .001. A third of patients improved in VA in both groups, 27% of patients gained more than 2 lines of vision in the CLRA-sparing group and 36% in the non-CLRA-sparing group. 17% improved to better than 20/200 in CLRA-sparing and 4% in non-CLRA sparing. Overall, 11% developed neovascularization all in non-CLRA sparing. In a multiple linear regression, VA at presentation was associated with regaining vision of 2 lines or more (OR = 2.603, P = .007). OCT showed progressive thinning over time, reaching lowest measurements at 6 months, and stabilizing thereafter. CONCLUSIONS In this modern cohort of acute CRAO patients, presentation to a tertiary facility within 12 hours of symptoms was seen in almost half of the patients. Final VA improved in almost a third of the patients, however, vision better than the legal blindness limit was rare (∼5%). Interestingly, a third of patients had some mild elevation of systemic inflammatory markers. Better VA at presentation was associated with visual gain, while baseline OCT values had poor correlation with final outcome.
Collapse
Affiliation(s)
- Sandra Hoyek
- From the Department of Ophthalmology, Massachusetts Eye and Ear (S.H., M.Y., R.B., J.W.M., D.G.V., and N.A.P.), Harvard Medical School, Boston, Massachusetts, USA
| | - Melissa Yuan
- From the Department of Ophthalmology, Massachusetts Eye and Ear (S.H., M.Y., R.B., J.W.M., D.G.V., and N.A.P.), Harvard Medical School, Boston, Massachusetts, USA
| | - Romy Bejjani
- From the Department of Ophthalmology, Massachusetts Eye and Ear (S.H., M.Y., R.B., J.W.M., D.G.V., and N.A.P.), Harvard Medical School, Boston, Massachusetts, USA
| | - David N Zacks
- Department of Ophthalmology, Kellogg Eye Center (D.N.Z.), University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Joan W Miller
- From the Department of Ophthalmology, Massachusetts Eye and Ear (S.H., M.Y., R.B., J.W.M., D.G.V., and N.A.P.), Harvard Medical School, Boston, Massachusetts, USA
| | - Demetrios G Vavvas
- From the Department of Ophthalmology, Massachusetts Eye and Ear (S.H., M.Y., R.B., J.W.M., D.G.V., and N.A.P.), Harvard Medical School, Boston, Massachusetts, USA
| | - Nimesh A Patel
- From the Department of Ophthalmology, Massachusetts Eye and Ear (S.H., M.Y., R.B., J.W.M., D.G.V., and N.A.P.), Harvard Medical School, Boston, Massachusetts, USA.
| |
Collapse
|
4
|
Han Y, Dai Y, Wang K, Zhang X, Shao Z, Zhu X. Post-pandemic insights on COVID-19 and premature ovarian insufficiency. Open Life Sci 2025; 20:20221028. [PMID: 39886482 PMCID: PMC11780258 DOI: 10.1515/biol-2022-1028] [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: 10/23/2024] [Revised: 11/25/2024] [Accepted: 11/28/2024] [Indexed: 02/01/2025] Open
Abstract
The COVID-19 pandemic has raised concerns regarding its potential impact on premature ovarian insufficiency (POI). This overview examines the possible interactions between COVID-19 and POI, while also suggesting preventive measures. The viral infection's inflammatory response and immune dysregulation may adversely affect ovarian tissues, leading to inflammation and damage. Additionally, alterations in vascular function could impair ovarian blood flow and hormonal imbalances may disrupt normal ovarian function. Long-term health effects, such as "long COVID," may exacerbate these issues through chronic inflammation and immune dysfunction. Public health measures, such as vaccination and home isolation, may indirectly protect ovarian health by reducing systemic inflammation. Vaccines could mitigate the severity of COVID-19's impact on ovarian function, while isolation may reduce stress and inflammation. However, further research is needed to validate these mechanisms.
Collapse
Affiliation(s)
- Yaguang Han
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Yang Dai
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Kexin Wang
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xin Zhang
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Zishen Shao
- Department of Medicine, First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Xiaolin Zhu
- Department of Medicine, Second Affiliated Hospital of Heilongjiang University of Chinese Medicine, No. 411 Guogeli Street, Nangang District, Harbin, Heilongjiang, 150006, P.R. China
| |
Collapse
|
5
|
Waclawovsky AJ, Dos Santos EB, de Oliveira AAR, Stubbs B, Schuch FB. Plasma biomarkers of endothelial function in people with depressive disorder: A systematic review and meta-analysis. J Affect Disord 2025; 375:297-305. [PMID: 39884367 DOI: 10.1016/j.jad.2025.01.138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 01/10/2025] [Accepted: 01/27/2025] [Indexed: 02/01/2025]
Abstract
The objective of this study is to conduct a literature review and summarize existing research comparing levels of blood markers of endothelial function in people with depression with controls. We searched major databases (Embase, PubMed, Web of Science, and PsycINFO) from inception to 23.07.2023, for studies comparing endothelial function blood markers in people with depression and controls including intercellular adhesion molecule-1 (sICAM-1), vascular cell adhesion molecule-1 (sVCAM-1), monocyte chemoattractant protein-1 (MCP-1), and sE-selectin. Comparative random effects meta-analysis, calculating standardized mean differences in levels of sICAM-1, sVCAM-1, MCP-1, and sE-selectin between depressed and controls were conducted. Study protocol was registered with PROSPERO (CRD42020222487). Nineteen studies including 10,566 participants [1322 depressed (mean age = 49.1 years, 63 % females) and 9239 controls (mean age = 52.3 years, 57.5 % females)] were included. People with depression had higher levels of sICAM-1 (SMD = 0.98, 95 % CI: 0.52; 1.44, P < 0.001) and MCP-1 (SMD = 0.37, 95 % CI: 0.01; 0.72, P = 0.04) than controls. There was a small difference on sVCAM-1 (k = 10, SMD = 0.215, 95%CI: 0.013; 0.417, p = 0.04), but no difference in levels of E-selectin between people with depression and controls (k = 5,SMD = 0.21, 95%CI:-0.09;0.52, P = 0.17). Subgroup analyzes demonstrated that people with depression and comorbid chronic illnesses, such as diabetes, have higher levels of sICAM-1 than people with depression without comorbid chronic illness. Mean age moderate the difference in sICAM-1 levels between depressed and controls (SMD = 0.01, 95%CI:-0.05;-0.02,P < 0.001, R2 = 0.61). Our findings suggest that people with depression exhibit increased levels of endothelial activation markers compared to controls, suggesting an endothelial dysfunction among this population.
Collapse
Affiliation(s)
- Aline Josiane Waclawovsky
- Postgraduate Program in Psychiatry and Mental Health, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Eduarda Bitencourt Dos Santos
- Postgraduate Program in Movement Sciences and Rehabilitation, Federal University of Santa Maria, Santa Maria, Brazil
| | | | - Brendon Stubbs
- Department of Psychological Medicine, Institute of Psychiatry, Psychology and Neuroscience (IoPPN), King's College London, London, UK; Centre for Sport Science and University Sports, University of Vienna, A- 1150 Wien, Austria
| | - Felipe Barreto Schuch
- Postgraduate Program in Psychiatry and Mental Health, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil; Postgraduate Program in Movement Sciences and Rehabilitation, Federal University of Santa Maria, Santa Maria, Brazil; Faculty of Health Sciences, Universidad Autónoma de Chile, Providencia, Chile
| |
Collapse
|
6
|
Abolfazli S, Karav S, Johnston TP, Sahebkar A. Regulatory effects of resveratrol on nitric oxide signaling in cardiovascular diseases. Pharmacol Rep 2025:10.1007/s43440-025-00694-w. [PMID: 39832074 DOI: 10.1007/s43440-025-00694-w] [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: 10/22/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Cardiovascular illnesses are multifactorial disorders and represent the primary reasons for death worldwide, according to the World Health Organization. As a signaling molecule, nitric oxide (NO) is extremely permeable across cellular membranes owing to its unique molecular features, like its small molecular size, lipophilicity, and free radical properties. Some of the biological effects of NO are vasodilation, inhibition in the growth of vascular smooth muscle cells, and functional regulation of cardiac cells. Several therapeutic approaches have been tested to increase the production of NO or some downstream NO signaling pathways. The health benefits of red wine are typically attributed to the polyphenolic phytoalexin, resveratrol (3,5,4'-trihydroxy-trans-stilbene), which is found in several plant species. Resveratrol has beneficial cardiovascular properties, some of which are mediated through endothelial nitric oxide synthase production (eNOS). Resveratrol promotes NO generation from eNOS through various methods, including upregulation of eNOS expression, activation in the enzymatic activity of eNOS, and reversal of eNOS uncoupling. Additionally, by reducing of oxidative stress, resveratrol inhibits the formation of superoxide and inactivation NO, increasing NO bioavailability. This review discusses the scientific literature on resveratrol's beneficial impact on NO signaling and how this effect improves the function of vascular endothelium.
Collapse
Affiliation(s)
- Sajad Abolfazli
- Student Research Committee, School of Pharmacy, Mazandaran University of Medical Science, Sari, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale, 17100, Turkey
| | - Thomas P Johnston
- Division of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
7
|
Kleeberg A, Luft T, Golkowski D, Purrucker JC. Endothelial dysfunction in acute ischemic stroke: a review. J Neurol 2025; 272:143. [PMID: 39812851 PMCID: PMC11735568 DOI: 10.1007/s00415-025-12888-6] [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: 11/06/2024] [Revised: 12/22/2024] [Accepted: 12/28/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND AND PURPOSE Endothelial dysfunction is considered an emerging therapeutic target to prevent complications during acute stroke and to prevent recurrent stroke. This review aims to provide an overview of the current knowledge on endothelial dysfunction, outline the diagnostic methods used to measure it and highlight the drugs currently being investigated for the treatment of endothelial dysfunction in acute ischemic stroke. METHODS The PubMed® and ClinicalTrials.gov electronic databases were searched for eligible articles/studies dealing with endothelial dysfunction and stroke. The references of the articles were screened to identify additional sources. The data were abstracted and summarized. FINDINGS AND DISCUSSION Endothelial dysfunction can be measured by serum biomarkers as well as by ultrasound or plethysmography techniques. Drugs targeting endothelial dysfunction include widely used agents such as angiotensin-converting enzyme inhibitors or isosorbide mononitrate, but also experimental therapies such as endothelial progenitor cells. CONCLUSION The role of endothelial dysfunction in acute ischemic stroke has been studied increasingly in recent years. It has been shown that there is a correlation between endothelial dysfunction and parenchymal hematoma after endovascular thrombectomy. Also, early clinical trials are conducted investigating, e.g., endothelial progenitor cells in the treatment of endothelial dysfunction in ischemic stroke. Current research focuses on the integration of novel markers of endothelial dysfunction into routine clinical practice to support decision making in the treatment of acute ischemic stroke.
Collapse
Affiliation(s)
- Antonia Kleeberg
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Thomas Luft
- Department of Oncology and Hematology, University Hospital Heidelberg, Heidelberg, Germany
| | - Daniel Golkowski
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| | - Jan C Purrucker
- Department of Neurology, Heidelberg University Hospital, Heidelberg, Germany
| |
Collapse
|
8
|
El-Sayed OS, Alnajjar AZ, Arafa A, Mohammed HE, Elettreby AM, Ibraheem S, Tawfik DE, Abdullah MAA, Tolba MA. Association between risk of ischemic stroke and liver enzymes levels: a systematic review and meta-analysis. BMC Neurol 2025; 25:18. [PMID: 39806288 PMCID: PMC11726974 DOI: 10.1186/s12883-024-03875-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 09/20/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Ischemic stroke is a major public health concern, contributing significantly to global morbidity and mortality. Recent studies have suggested that alterations in liver enzymes may be linked to the risk of developing a stroke. However, the relationship between liver enzymes and ischemic stroke remains unclear. OBJECTIVE To examine the potential role of liver enzymes as biomarkers for ischemic stroke. METHODS We systematically searched four databases for articles investigating the association between liver enzymes and ischemic stroke up to March 20th, 2024. Newcastle Ottawa Scale judged the quality of included studies. Risk ratio (RR), hazard ratio (HR), or odds ratio (OR) were extracted and statistically analyzed by RevMan and R software. The Grading of Recommendations, Assessment, Development, and Evaluation (GRADE) assessed the certainty of evidence. RESULTS Increased levels of gamma-glutamyl transferase (GGT) and alkaline phosphatase (ALP) have shown statistically significant association with increased ischemic stroke risk (RR: 1.43, 95% CI: [1.30 to 1.57], P > 0.00001) and (RR: 1.60, 95% CI: [1.22 to 2.10], P = 0.0006), respectively. Conversely, increased levels of alanine aminotransferase (ALT) and aspartate aminotransferase (AST) showed no significant association with ischemic stroke risk (RR: 0.92, 95% CI: [0.68 to 1.24], P = 0.58) and (RR: 1.43, 95% CI: [0.83 to 2.49], P = 0.20), respectively. The evidence for all outcomes had a low or very low level of certainty. CONCLUSION GGT and ALP could be potential biomarkers for increased ischemic stroke risk, which necessitates careful follow-up. However, AST and ALT did not show such association.
Collapse
|
9
|
Grodecki K, Geers J, Kwiecinski J, Lin A, Slipczuk L, Slomka PJ, Dweck MR, Nerlekar N, Williams MC, Berman D, Marwick T, Newby DE, Dey D. Phenotyping atherosclerotic plaque and perivascular adipose tissue: signalling pathways and clinical biomarkers in atherosclerosis. Nat Rev Cardiol 2025:10.1038/s41569-024-01110-1. [PMID: 39743563 DOI: 10.1038/s41569-024-01110-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/20/2024] [Indexed: 01/04/2025]
Abstract
Computed tomography coronary angiography provides a non-invasive evaluation of coronary artery disease that includes phenotyping of atherosclerotic plaques and the surrounding perivascular adipose tissue (PVAT). Image analysis techniques have been developed to quantify atherosclerotic plaque burden and morphology as well as the associated PVAT attenuation, and emerging radiomic approaches can add further contextual information. PVAT attenuation might provide a novel measure of vascular health that could be indicative of the pathogenetic processes implicated in atherosclerosis such as inflammation, fibrosis or increased vascularity. Bidirectional signalling between the coronary artery and adjacent PVAT has been hypothesized to contribute to coronary artery disease progression and provide a potential novel measure of the risk of future cardiovascular events. However, despite the development of more advanced radiomic and artificial intelligence-based algorithms, studies involving large datasets suggest that the measurement of PVAT attenuation contributes only modest additional predictive discrimination to standard cardiovascular risk scores. In this Review, we explore the pathobiology of coronary atherosclerotic plaques and PVAT, describe their phenotyping with computed tomography coronary angiography, and discuss potential future applications in clinical risk prediction and patient management.
Collapse
Affiliation(s)
- Kajetan Grodecki
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
- 1st Department of Cardiology, Medical University of Warsaw, Warsaw, Poland
| | - Jolien Geers
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
- Department of Cardiology, Centrum Voor Hart- en Vaatziekten (CHVZ), Universitair Ziekenhuis Brussel (UZ Brussel), Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Jacek Kwiecinski
- Department of Interventional Cardiology and Angiology, National Institute of Cardiology, Warsaw, Poland
| | - Andrew Lin
- Monash Victorian Heart Institute and Monash Health Heart, Monash University, Victorian Heart Hospital, Melbourne, Victoria, Australia
| | - Leandro Slipczuk
- Division of Cardiology, Montefiore Healthcare Network/Albert Einstein College of Medicine, New York, NY, USA
| | - Piotr J Slomka
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
| | - Marc R Dweck
- British Heart Foundation Centre of Research Excellence, University of Edinburgh, Edinburgh, UK
| | - Nitesh Nerlekar
- Monash Victorian Heart Institute and Monash Health Heart, Monash University, Victorian Heart Hospital, Melbourne, Victoria, Australia
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Michelle C Williams
- British Heart Foundation Centre of Research Excellence, University of Edinburgh, Edinburgh, UK
| | - Daniel Berman
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA
| | - Thomas Marwick
- Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - David E Newby
- British Heart Foundation Centre of Research Excellence, University of Edinburgh, Edinburgh, UK
| | - Damini Dey
- Department of Biomedical Sciences, and Department of Medicine, Cedars-Sinai Medical Center, Biomedical Imaging Research Institute, Los Angeles, CA, USA.
| |
Collapse
|
10
|
Lu W, Teoh A, Waters M, Haug G, Shakeel I, Hassan I, Shahzad AM, Callerfelt AKL, Piccari L, Sohal SS. Pathology of idiopathic pulmonary fibrosis with particular focus on vascular endothelium and epithelial injury and their therapeutic potential. Pharmacol Ther 2025; 265:108757. [PMID: 39586361 DOI: 10.1016/j.pharmthera.2024.108757] [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: 10/15/2024] [Accepted: 11/20/2024] [Indexed: 11/27/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) remains a challenging disease with no drugs available to change the trajectory. It is a condition associated with excessive and highly progressive scarring of the lungs with remodelling and extracellular matrix deposition. It is a highly "destructive" disease of the lungs. The diagnosis of IPF is challenging due to continuous evolution of the disease, which also makes early interventions very difficult. The role of vascular endothelial cells has not been explored in IPF in great detail. We do not know much about their contribution to arterial or vascular remodelling, extracellular matrix changes and contribution to pulmonary hypertension and lung fibrosis in general. Endothelial to mesenchymal transition appears to be central to such changes in IPF. Similarly, for epithelial changes, the process of epithelial to mesenchymal transition seem to be the key both for airway epithelial cells and type-2 pneumocytes. We focus here on endothelial and epithelial cell changes and its contributions to IPF. In this review we revisit the pathology of IPF, mechanistic signalling pathways, clinical definition, update on diagnosis and new advances made in treatment of this disease. We discuss ongoing clinical trials with mode of action. A multidisciplinary collaborative approach is needed to understand this treacherous disease for new therapeutic targets.
Collapse
Affiliation(s)
- Wenying Lu
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Alan Teoh
- National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia
| | - Maddison Waters
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania 7250, Australia
| | - Greg Haug
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; Department of Respiratory Medicine, Launceston General Hospital, Launceston, Tasmania 7250, Australia
| | - Ilma Shakeel
- Centre For Interdisciplinary Research In Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Imtaiyaz Hassan
- Centre For Interdisciplinary Research In Basic Sciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Affan Mahmood Shahzad
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; Medical School, Oceania University of Medicine, Apia, Samoa
| | | | - Lucilla Piccari
- Department of Pulmonology, Hospital del Mar, Barcelona, Spain
| | - Sukhwinder Singh Sohal
- Respiratory Translational Research Group, Department of Laboratory Medicine, School of Health Sciences, University of Tasmania, Newnham, Tasmania 7248, Australia; National Health and Medical Research Council (NHMRC) Centre of Research Excellence (CRE) in Pulmonary Fibrosis, Respiratory Medicine and Sleep Unit, Royal Prince Alfred Hospital, Camperdown, NSW 2050, Australia.
| |
Collapse
|
11
|
Yu SM, Liu JQ, Zhang LL, Ma YT, Yin FY, Liu S. Mmu_circ_0001148 promotes endothlial-mesenchymal transition via regulating miR-218-5p/JMY axis and drives progression of atherosclerosis. Int J Biol Macromol 2024; 293:139305. [PMID: 39736291 DOI: 10.1016/j.ijbiomac.2024.139305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 12/26/2024] [Accepted: 12/27/2024] [Indexed: 01/01/2025]
Abstract
Atherosclerosis (AS) is a common cardiovascular disease and responsible for the high mortality of cardiovascular emergencies. Circular RNAs (circRNAs) show a potential role in atherogenesis. We identified an aberrantly expressed circRNA (circ_0001148) in atherosclerotic tissues. However, whether circ_0001148 involved in atherogenesis remains unclear. The present study aimed to investigate the biological function of circ_0001148 and the underlying mechanism in AS. Functional analysis indicated that circ_0001148 promoted endothelial-mesenchymal transition (EndMT) and thus accelerated the formation of atherosclerotic plaque. The mechanism analysis suggested that circ_0001148 act as a competitive endogenous RNA (ceRNA) to modify the expression of JMY by sponging miR-218-5p. We also demonstrated that the treatment of miR-218-5p mimics or JMY deficiency could attenuated the progression of AS induced by circ_0001148 overexpression. Therefore, we proposed a novel signaling network which circ_0001148 promotes atherogenesis via miR-218-5p/JMY axis. These findings provide new insights into the mechanisms of AS, and potentially leading to the development of a novel therapeutic strategy targeting circ_0001148.
Collapse
Affiliation(s)
- Shang-Min Yu
- Department of Pharmaceutics, School of Pharmacy, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Jia-Qi Liu
- School of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Lin-Lin Zhang
- Department of Physiology, School of Basic Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Ya-Ting Ma
- Department of Pharmaceutics, School of Pharmacy, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Fei-Yang Yin
- School of Clinical Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China
| | - Shan Liu
- Department of Physiology, School of Basic Medicine, Bengbu Medical University, Bengbu 233000, Anhui, China; Key Laboratory of Basic and Clinical Cardiovascular Diseases, Bengbu Medical University, Bengbu 233000, Anhui, China.
| |
Collapse
|
12
|
Wang Y, Cao L, Wang K, Chen J, Li X, Zhao Z, Han X, Ni K, Liu D, Wu X, Wang G. The IL-1β/STAT1 Axis inhibits STAT3 function via Sequestration of the transcriptional activator GLIS2, leading to postoperative vascular dysfunction. Int Immunopharmacol 2024; 143:113372. [PMID: 39418736 DOI: 10.1016/j.intimp.2024.113372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/01/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024]
Abstract
Surgery-induced endothelial dysfunction is crucial in thrombus formation, driven by the release of inflammatory mediators due to surgical trauma. The STAT family, known for amplifying inflammatory responses via cytokine activation, plays an unclear role in the signaling mechanisms from surgery to molecular activation, and their regulatory effects on inflammation vary. This study aimed to identify key signaling pathways responsible for vascular dysfunction post-surgery and to discover potential targets for predicting or preventing thrombosis. To explore this, endothelial cells were co-cultured with post-surgical trauma serum and analyzed using various assays. Bioinformatics analysis linked surgical trauma with pathways involving thrombosis, interleukins, cytokines, and STAT signaling. Elevated inflammatory mediators were observed in mouse serum post-surgical trauma, with IL-6 activating STAT3 to enhance endothelial proliferation, while IL-1β activated STAT1, inhibiting STAT3's effects. Gli-similar 2 (GLIS2), a novel coactivator of STAT3, was found to regulate STAT transcription. STAT1, however, inhibited GLIS2's interaction with STAT3, suppressing STAT3's role in endothelial proliferation. The study concludes that IL-1β-triggered STAT1 activation impedes GLIS2-STAT3 interaction, reducing STAT3's transcriptional activity and leading to endothelial dysfunction, presenting new targets for preventing post-surgical trauma endothelial dysfunction and thrombosis.
Collapse
Affiliation(s)
- Yi Wang
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Liang Cao
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ke Wang
- Department of Neonatal Medical Center, Qingdao Women and Children's Hospital, Qingdao University, Class B Key Disciplines of Newborns, Qingdao 266000, China
| | - Jing Chen
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin 150081, Heilongjiang, China
| | - Xinxin Li
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Zinan Zhao
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Xue Han
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China
| | - Ke Ni
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin 150081, Heilongjiang, China
| | - Dandan Liu
- Department of Anesthesiology, Harbin Medical University Cancer Hospital, Haping Road No. 150, Nangang District, Harbin 150081, Heilongjiang, China
| | - Xiaohong Wu
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| | - Guonian Wang
- Department of Anesthesiology, the Fourth Affiliated Hospital of Harbin Medical University, Harbin 150001, China.
| |
Collapse
|
13
|
Luo X, Zhang S, Wang L, Li J. Pathological roles of mitochondrial dysfunction in endothelial cells during the cerebral no-reflow phenomenon: A review. Medicine (Baltimore) 2024; 103:e40951. [PMID: 39705421 DOI: 10.1097/md.0000000000040951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2024] Open
Abstract
Emergency intravascular interventional therapy is the most effective approach to rapidly restore blood flow and manage occlusion of major blood vessels during the initial phase of acute ischemic stroke. Nevertheless, several patients continue to experience ineffective reperfusion or cerebral no-reflow phenomenon, that is, hypoperfusion of cerebral blood supply after treatment. This is primarily attributed to downstream microcirculation disturbance. As integral components of the cerebral microvascular structure, endothelial cells (ECs) attach importance to regulating microcirculatory blood flow. Unlike neurons and microglia, ECs harbor a relatively low abundance of mitochondria, acting as key sensors of environmental and cellular stress in regulating the viability, structural integrity, and function of ECs rather than generating energy. Mitochondria dysfunction including increased mitochondrial reactive oxygen species levels and disturbed mitochondrial dynamics causes endothelial injury, further causing microcirculation disturbance involved in the cerebral no-reflow phenomenon. Therefore, this review aims to discuss the role of mitochondrial changes in regulating the role of ECs and cerebral microcirculation blood flow during I/R injury. The outcomes of the review will provide promising potential therapeutic targets for future prevention and effective improvement of the cerebral no-reflow phenomenon.
Collapse
Affiliation(s)
- Xia Luo
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Shaotao Zhang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Longbing Wang
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinglun Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Neurological Diseases and Brain Function, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
14
|
Vakili S, Cao K. Angiopoietin-2: A Therapeutic Target for Vascular Protection in Hutchinson-Gilford Progeria Syndrome. Int J Mol Sci 2024; 25:13537. [PMID: 39769300 PMCID: PMC11676795 DOI: 10.3390/ijms252413537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a pediatric condition characterized by clinical features that resemble accelerated aging. The abnormal accumulation of a toxic form of the lamin A protein known as progerin disrupts cellular functions, leading to various complications, including growth retardation, loss of subcutaneous fat, abnormal skin, alopecia, osteoporosis, and progressive joint contractures. Death primarily occurs as the result of complications from progressive atherosclerosis, especially from cardiac disease, such as myocardial infarction or heart failure, or cerebrovascular disease like stroke. Despite the availability of lonafarnib, the only US Food and Drug Administration-approved treatment for HGPS, cardiovascular complications remain the leading cause of morbidity and mortality in affected patients. Defective angiogenesis-the process of forming new blood vessels from existing ones-plays a crucial role in the development of cardiovascular disease. A recent study suggests that Angiopoietin-2 (Ang2), a pro-angiogenic growth factor that regulates angiogenesis and vascular stability, may offer therapeutic potential for the treatment of HGPS. In this review, we describe the clinical features and key cellular processes impacted by progerin and discuss the therapeutic potential of Ang2 in addressing these challenges.
Collapse
Affiliation(s)
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742, USA;
| |
Collapse
|
15
|
Alkharfy KM, Ahmad A, Almuaijel S, Bin Hashim A, Raish M, Jan BL, Rehman NU, Anwar F, Rehman MT, Alajmi MF. The vascular effects of peppermint ( Mentha longifolia. L) on aorta in a mouse model: an ex-vivo and computational study. J Biomol Struct Dyn 2024:1-16. [PMID: 39663630 DOI: 10.1080/07391102.2024.2439616] [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: 12/18/2023] [Accepted: 04/24/2024] [Indexed: 12/13/2024]
Abstract
The present study examined the vascular effects of peppermint or mint (Mentha longifolia L.) using an abdominal aortic rings model. Concentration-response curves for mint oil were generated after precontracting isolated mouse aorta with phenylephrine. The effect of different receptor antagonists and ion channel or enzyme inhibitors on the vasorelaxant potential of mint oil were studied. Molecular docking studies were conducted using computational techniques to investigate the potential interactions between the bioactive constituents of mint oil and key vascular targets. The tension of aortic rings, which had been contracted by phenylephrine, relaxed as a function of the concentration of mint oil (0.0002-2 mg/mL). Pretreatment of the rings with the nitric oxide synthase inhibitor (L-NAME), a nonselective β-blocker (propranolol), and a muscarinic receptor blocker (atropine) didn't show significant resistance to the vasodilatory effects of the mint oil. The vasodilatory effects of mint oil were significantly diminished when the rings were pretreated with glibenclamide, an inhibitor of ATP-sensitive K+ channels. In addition, indomethacin, a cyclooxygenase (COX) inhibitor, did influence mint oil's tension in the preparations precontracted with phenylephrine. The present findings imply that ATP-sensitive K+ channels activation, blocking of Ca2+ channels, and inhibition of COX play a role in mediating the mint oil-induced vasorelaxation. Molecular docking studies of mint oil constituents showed that β-Elemene and Aromadendrene can interact with K+ and Ca2+ channels through various hydrophobic interactions with key amino acid residues. Additional work is needed to confirm the possible beneficial application of mint oil or its constituents in regulating the vascular tone.
Collapse
Affiliation(s)
- Khalid M Alkharfy
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Ajaz Ahmad
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Saleh Almuaijel
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah Bin Hashim
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammad Raish
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Basit L Jan
- Department of Clinical Pharmacy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Najeeb Ur Rehman
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Saudi Arabia
| | - Farooq Anwar
- Department of Food Sciences, Faculty of Food Sciences and Technology, Universiti Putra Malaysia 43400, Serdang, Malaysia
- Institute of Chemistry, University of Sargodha, Sargodha, Pakistan
| | - Md Tabish Rehman
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohamad F Alajmi
- Department of Pharmacognosy, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
16
|
Lu QY, Guo L, Zhang QY, Yang FM, Zhou ST, Sun QY. Luteolin Alleviates the TNF- α-Induced Inflammatory Response of Human Microvascular Endothelial Cells via the Akt/MAPK/NF- κB Pathway. Mediators Inflamm 2024; 2024:6393872. [PMID: 39698583 PMCID: PMC11655144 DOI: 10.1155/mi/6393872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/20/2024] Open
Abstract
Endothelial dysfunction and pathological alterations are pivotal in the pathogenesis of cardiovascular disease. To date, effective interventions for these endothelial changes are lacking. Tumor necrosis factor-alpha (TNF-α) is known to significantly contribute to these alterations. It has been reported the potential of luteolin to mitigate TNF-α-induced inflammation, yet its specific mechanisms and targets still remain to be elucidated. This study aims to investigate the effects and mechanisms of luteolin on TNF-α-induced inflammatory injury in human microvascular endothelial cells, thereby advancing the understanding of luteolin's medicinal properties. Our findings demonstrate that luteolin notably inhibits TNF-α-induced phosphorylation of Akt, mitogen activated protein kinase (MAPK), and the nuclear factor-kappaB (NF-κB) p65. It significantly reduces the transcriptional activity of NF-κB p65 and AP-1 and decreases the expression of mRNA and proteins related to adhesion molecules and inflammatory mediators. Additionally, luteolin inhibited the reduction in STAT3 phosphorylation. In conclusion, luteolin effectively suppresses TNF-α-induced inflammatory injury in endothelial cells via the Akt/MAPK/NF-κB pathway.
Collapse
Affiliation(s)
- Qing-Yu Lu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Institute of Pharmacology and Bioactivity, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Li Guo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Institute of Pharmacology and Bioactivity, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Qi-Yun Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Institute of Pharmacology and Bioactivity, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Fu-Mei Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Institute of Pharmacology and Bioactivity, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Shu-Ting Zhou
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Institute of Pharmacology and Bioactivity, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| | - Qian-Yun Sun
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China
- Institute of Pharmacology and Bioactivity, Natural Products Research Center of Guizhou Province, Guiyang 550014, China
| |
Collapse
|
17
|
Cholidis P, Kranas D, Chira A, Galouni EA, Adamantidi T, Anastasiadou C, Tsoupras A. Shrimp Lipid Bioactives with Anti-Inflammatory, Antithrombotic, and Antioxidant Health-Promoting Properties for Cardio-Protection. Mar Drugs 2024; 22:554. [PMID: 39728129 DOI: 10.3390/md22120554] [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: 10/15/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 12/28/2024] Open
Abstract
Marine animals, especially shrimp species, have gained interest in research, due to the fact that they contain a plethora of biomolecules, specifically lipids, which have been proven to possess many health benefits in various diseases linked to chronic inflammation or other exogenous factors. This review refers to the lipid composition of a large number of shrimp species, as well as the effects that can alternate the lipid content of these crustaceans. Emphasis is given to the potent anti-inflammatory, antioxidant, and antithrombotic properties of shrimp bioactives, as well as the effects that these bioactives hold in other diseases, such as cancer, diabetes, neurodegenerative disorders, and more. The various health-promoting effects deriving from the consumption of shrimp lipid bioactives and the usage of products containing shrimp lipid extracts are also addressed in this study, through the exploration of several mechanisms of action and the interference of shrimp lipids in these biochemical pathways. Nevertheless, further research on this cultivatable edible species is needed, due to their existing limitations and future prospects which are discussed in this paper.
Collapse
Affiliation(s)
- Paschalis Cholidis
- Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
| | - Dimitrios Kranas
- Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
| | - Aggeliki Chira
- Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
| | - Evangelia Aikaterini Galouni
- Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
| | - Theodora Adamantidi
- Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
| | | | - Alexandros Tsoupras
- Hephaestus Laboratory, School of Chemistry, Faculty of Sciences, Democritus University of Thrace, Kavala University Campus, St. Lucas, 65404 Kavala, Greece
| |
Collapse
|
18
|
Fakir S, Kubra KT, Akhter MS, Uddin MA, Sarker MMR, Siejka A, Barabutis N. Unfolded protein response modulates the effects of GHRH antagonists in experimental models of in vivo and in vitro lung injury. Tissue Barriers 2024:2438974. [PMID: 39653654 DOI: 10.1080/21688370.2024.2438974] [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: 10/18/2024] [Revised: 11/29/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024] Open
Abstract
The development of efficient targeted therapies to ameliorate endothelial disorders is of the utmost need, as evident by the devastating outcomes of the recent pandemic. Recent findings suggest that unfolded protein response (UPR) modulates barrier function. In the current study, we reveal that the aforementioned highly conservative mechanism is involved in the protective effects of growth hormone-releasing hormone antagonists (GHRHAnt) in lung injury, both in vivo and in vitro. In bovine pulmonary artery endothelial cells, UPR suppression counteracted the protective effects of GHRHAnt in lipopolysaccharide (LPS)-induced endothelial hyperpermeability. In mouse lungs, UPR activation enhanced the beneficial effects of GHRHAnt against LPS-induced acute lung injury. Our observations - which are focused on lung endothelial cells and tissues - enhance our knowledge on the mechanisms mediating the barrier function and contribute to the development of novel therapies toward sepsis, direct and indirect lung injury. The effects of UPR modulation on the effects of GHRHAnt in other tissues are unknown, and they are the subject of future investigations.
Collapse
Affiliation(s)
- Saikat Fakir
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Khadeja-Tul Kubra
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Mohammad Shohel Akhter
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Mohammad Afaz Uddin
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Md Matiur Rahman Sarker
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| | - Agnieszka Siejka
- Department of Clinical Endocrinology, Medical University of Lodz, Lodz, Poland
| | - Nektarios Barabutis
- School of Basic Pharmaceutical and Toxicological Sciences, College of Pharmacy, University of Louisiana Monroe, Monroe, Louisiana, USA
| |
Collapse
|
19
|
Dobariya KH, Goyal D, Kumar H. Molecular signature-based labeling techniques for vascular endothelial cells. Acta Histochem 2024; 127:152222. [PMID: 39644518 DOI: 10.1016/j.acthis.2024.152222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 11/14/2024] [Accepted: 11/26/2024] [Indexed: 12/09/2024]
Abstract
Vascular endothelial cells (VECs) play a crucial role in the development and maintenance of vascular biology specific to the tissue types. Molecular signature-based labeling and imaging of VECs help researchers understand potential mechanisms linking VECs to disease pathology, serving as valuable biomarkers in clinical settings and trials. Labeling techniques involve selectively tagging or marking VECs for visualization. Immunolabeled employs antibodies that specifically bind to VECs markers, while fluorescent tracers or dyes can directly label VECs for imaging. Some techniques use specific carbohydrate residues on cell surface, while others employ endothelial-specific promoters to express fluorescent proteins. Additionally, VEC can be labeled with contrast agents, radiolabeled tracers, and nanoparticles. The choice of labeling technique depends on study context, including whether it involves animal models, in vitro cell cultures, or clinical applications. Herein, we discussed the various labeling methods utilized to label VECs and the techniques to visualize them.
Collapse
Affiliation(s)
- Krutika H Dobariya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Divya Goyal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Hemant Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
20
|
Amin T, Rasool MHU, Ozkan BI, Swaminathan G, Rauf F, Patrizi S, Sethi A, Frishman WH, Aronow WS, Ahmed MS. Leukocytosis as a Risk Factor for Coronary Artery Disease: Pathophysiology and Epidemiology. Cardiol Rev 2024. [DOI: 10.1097/crd.0000000000000824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Coronary artery disease (CAD) is a significant health concern characterized by reduced blood flow to the heart muscle, primarily due to the buildup of atherosclerotic plaques in the coronary arteries. This process begins with endothelial injury, leading to a cascade of biological responses contributing to plaque formation. Endothelial injury attracts the migration of monocytes which differentiate into macrophages upon uptake of oxidized low-density lipoproteins, changing into lipid-laden macrophage or “foam cells.” The process of plaque formation is influenced by many factors which have been studied extensively in literature such as smoking, hypertension, and diabetes mellitus. Chronic inflammatory illnesses are often associated with a high prevalence of coronary artery syndromes, prompting the evaluation of markers of inflammation such as white blood cell count and inflammatory markers as independent risk factors for CAD. White blood cells play a remarkable role in the pathophysiology of disease formation and progression. The article below aims to discuss the pathophysiology and epidemiology of leukocytosis as a risk factor for CAD.
Collapse
Affiliation(s)
- Toka Amin
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | | | - Bike Ilyada Ozkan
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Gowri Swaminathan
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Faateh Rauf
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Santino Patrizi
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | - Arshia Sethi
- Department of Medicine, Icahn School of Medicine at Mount Sinai Medical Center, New York, NY
| | | | - Wilbert S. Aronow
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| | - Mahmoud Samy Ahmed
- Departments of Cardiology and Medicine, Westchester Medical Center and New York Medical College, Valhalla, NY
| |
Collapse
|
21
|
Hasan ML, Lee JR, Rahaman KA, Yang DH, Joung YK. Versatile effects of galectin-1 protein-containing lipid bilayer coating for cardiovascular applications. Bioact Mater 2024; 42:207-225. [PMID: 39285911 PMCID: PMC11403261 DOI: 10.1016/j.bioactmat.2024.08.026] [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: 05/27/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/19/2024] Open
Abstract
Modulating inflammatory cells in an implantation site leads to severe complications and still unsolved challenges for blood-contacting medical devices. Inspired by the role of galectin-1 (Gal-1) in selective functions on multiple cells and immunomodulatory processes, we prepared a biologically target-specific surface coated with the lipid bilayer containing Gal-1 (Gal-1-SLB) and investigate the proof of the biological effects. First, lipoamido-dPEG-acid was deposited on a gold-coated substrate to form a self-assembled monolayer and then conjugated dioleoylphosphatidylethanolamine (DOPE) onto that to produce a lower leaflet of the supported lipid bilayer (SLB) before fusing membrane-derived vesicles extracted from B16-F10 cells. The Gal-1-SLB showed the expected anti-fouling activity by revealing the resistance to protein adsorption and bacterial adhesion. In vitro studies showed that the Gal-1-SLB can promote endothelial function and inhibit smooth muscle cell proliferation. Moreover, Gal-1- SLB presents potential function for endothelial cell migration and angiogenic activities. In vitro macrophage culture studies showed that the Gal-1-SLB attenuated the LPS-induced inflammation and the production of macrophage-secreted inflammatory cytokines. Finally, the implanted Gal-1-SLB reduced the infiltration of immune cells at the tissue-implant interface and increased markers for M2 polarization and blood vessel formation in vivo. This straightforward surface coating with Gal-1 can be a useful strategy for modulating the vascular and immune cells around a blood-contacting device.
Collapse
Affiliation(s)
- Md Lemon Hasan
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
| | - Ju Ro Lee
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Center for Systems Biology, Massachusetts General Hospital Research Institute, Boston, MA, 02114, USA
- Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, 02114, USA
| | - Khandoker Asiqur Rahaman
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
| | - Dae Hyeok Yang
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Yoon Ki Joung
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarangno 14-gil 5, Seongbuk-gu, Seoul, 02792, Republic of Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), 113 Gwahangno, Yuseong-gu, Daejeon, 34113, Republic of Korea
- KHU-KIST Department of Conversing Science and Technology, Graduate School, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
22
|
Hannawi Y. Cerebral Small Vessel Disease: a Review of the Pathophysiological Mechanisms. Transl Stroke Res 2024; 15:1050-1069. [PMID: 37864643 DOI: 10.1007/s12975-023-01195-9] [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: 06/02/2023] [Revised: 06/02/2023] [Accepted: 09/18/2023] [Indexed: 10/23/2023]
Abstract
Cerebral small vessel disease (cSVD) refers to the age-dependent pathological processes involving the brain small vessels and leading to vascular cognitive impairment, intracerebral hemorrhage, and acute lacunar ischemic stroke. Despite the significant public health burden of cSVD, disease-specific therapeutics remain unavailable due to the incomplete understanding of the underlying pathophysiological mechanisms. Recent advances in neuroimaging acquisition and processing capabilities as well as findings from cSVD animal models have revealed critical roles of several age-dependent processes in cSVD pathogenesis including arterial stiffness, vascular oxidative stress, low-grade systemic inflammation, gut dysbiosis, and increased salt intake. These factors interact to cause a state of endothelial cell dysfunction impairing cerebral blood flow regulation and breaking the blood brain barrier. Neuroinflammation follows resulting in neuronal injury and cSVD clinical manifestations. Impairment of the cerebral waste clearance through the glymphatic system is another potential process that has been recently highlighted contributing to the cognitive decline. This review details these mechanisms and attempts to explain their complex interactions. In addition, the relevant knowledge gaps in cSVD mechanistic understanding are identified and a systematic approach to future translational and early phase clinical research is proposed in order to reveal new cSVD mechanisms and develop disease-specific therapeutics.
Collapse
Affiliation(s)
- Yousef Hannawi
- Division of Cerebrovascular Diseases and Neurocritical Care, Department of Neurology, The Ohio State University, 333 West 10th Ave, Graves Hall 3172C, Columbus, OH, 43210, USA.
| |
Collapse
|
23
|
Yilmaz E, Yilmaz D, Cacan E. Severe and post-COVID-19 are associated with high expression of vimentin and reduced expression of N-cadherin. Sci Rep 2024; 14:29256. [PMID: 39587116 PMCID: PMC11589739 DOI: 10.1038/s41598-024-72192-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/04/2024] [Indexed: 11/27/2024] Open
Abstract
SARS-CoV-2 penetrates human cells via its spike protein, which mainly interacts with ACE2 receptors, triggering viral replication and an exacerbated immune response characterized by a cytokine storm. Vimentin III, an intermediate filament protein predominantly found in mesenchymal cells, has garnered considerable attention in recent research due to its multifaceted biological roles and significance in the endothelial-mesenchymal transition (EndMT) during various fibrotic processes. However, the pathophysiological mechanisms linking vimentin to SARS-CoV-2 remain incompletely elucidated. In this study, we determined the expression profiles of vimentin in three cohorts: patients admitted to the intensive care unit with SARS-CoV-2 infection, individuals in the 6-12 month convalescent phase post-infection and COVID-19 negative controls. Our objective was to assess the association between peripheral blood biomarkers implicated in endothelial dysfunction and genes related to fibrosis. Serum levels of vimentin and N-cadherin were determined by ELISA, while vimentin gene expression was determined by qRT-PCR. In addition, we examined the correlation between clinical parameters and serum levels of vimentin and N-cadherin in severe COVID-19 patients and healthy counterparts. Our findings revealed elevated serum vimentin levels and increased gene expression in severe COVID-19 patients compared to healthy controls. Conversely, serum N-cadherin levels were diminished in both acute and convalescent stages of severe COVID-19 relative to healthy individuals. Notably, associations were observed between C-reactive protein, lactate dehydrogenase, lymphocyte count and vimentin levels in severe COVID-19 patients, indicative of endothelial dysfunction. Furthermore, our study identified vimentin and N-cadherin as potential diagnostic markers via ROC analysis. Overall, delineating the dysregulation of vimentin and N-cadherin due to SARS-CoV-2 infection in disease pathogenesis and tissue homeostasis offers novel insights for clinical management and targeted therapeutic interventions.
Collapse
Affiliation(s)
- Esra Yilmaz
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Tokat Gaziosmanpasa University, 60200, Tokat, Turkey
| | - Dilek Yilmaz
- Department of Infectious Diseases and Clinical Microbiology, Yozgat City Hospital, 66100, Yozgat, Turkey
| | - Ercan Cacan
- Department of Molecular Biology and Genetics, Faculty of Arts and Sciences, Tokat Gaziosmanpasa University, 60200, Tokat, Turkey.
| |
Collapse
|
24
|
Gao Y, Chen H, Ke D, Li C, Jiang Z, Pu B. Effect of muscle strength on deep vein thrombosis: A Mendelian randomization study. Medicine (Baltimore) 2024; 103:e40138. [PMID: 39533595 PMCID: PMC11557092 DOI: 10.1097/md.0000000000040138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Deep vein thrombosis (DVT) is a serious condition characterized by blood clots in deep veins, posing a significant public health burden. Muscle strength has been implicated as a potential risk factor for DVT due to its influence on venous return. This study aims to investigate the causal association between muscle strength and DVT using a Mendelian randomization (MR) approach, leveraging genetic variants as instrumental variables (IVs). We conducted a 2-sample MR analysis using genome-wide association study (GWAS) data for hand-grip strength and DVT. IVs were selected based on their significant associations with muscle strength and DVT, as well as their linkage disequilibrium patterns. We employed statistical methods including inverse-variance weighting (IVW), MR-Egger, and weighted median to address pleiotropy bias. Sensitivity analyses were conducted to evaluate the robustness of the results. A total of 21 and 14 independent IVs were identified for hand grip strength (EWGSOP) and hand grip strength (FNIH), respectively. IVW analysis revealed a consistent causal and negative association between both definitions of hand grip strength and DVT (EWGSOP: OR = 0.702, 95% CI: 0.511-0.964, P = .029; FNIH: OR = 0.715, 95% CI: 0.570-0.898, P = .004). No directional pleiotropy was detected in MR-Egger and MR-PRESSO analyses for either definition (EWGSOP: MR-Egger Intercept P = .516; MR-PRESSO global test P = .162; FNIH: MR-Egger Intercept P = .569; MR-PRESSO global test P = .371).Sensitivity analyses demonstrated the stability of the causal effect estimates, with little influence from individual IVs. The MR analysis provided evidence of a causal association between muscle strength and DVT risk, suggesting that increasing muscle strength may have a protective effect. These findings have implications for preventive strategies and the promotion of resistance exercises and muscle-strengthening activities. Further research and validation of these results could inform clinical guidelines and interventions for DVT prevention.
Collapse
Affiliation(s)
- Yijia Gao
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Hui Chen
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Daoze Ke
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Changfan Li
- Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Ziwei Jiang
- The First Affiliated Hospital, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong Province, China
| | - Bin Pu
- Department of Orthopedics, Suining Traditional Chinese Medicine Hospital Affiliated to North Sichuan Medical College, Suining, Sichuan Province, China
| |
Collapse
|
25
|
Chen D, Xu W, Zheng H, Zhang Y, Lin Y, Han Y, Yao F, Shen H. The methyltransferase METTL3 regulates endothelial cell proliferation and inflammation via m 6A RNA methylation-mediated TRAF1 expression. Biochem Biophys Res Commun 2024; 732:150399. [PMID: 39033551 DOI: 10.1016/j.bbrc.2024.150399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
The imbalance of vascular endothelial cell homeostasis is the key mechanism for the progression of many vascular diseases. RNA modification, particularly N6-Methyladenosine (m6A), plays important function in numerous biological processes. Nevertheless, the regulatory function of m6A RNA methylation in endothelial dysfunction remains insufficiently characterized. In this study, we established that the m6A methyltransferase METTL3 is critical for regulating endothelial function. Functionally, depletion of METTL3 results in decreased endothelial cells proliferation, survival and inflammatory response. Conversely, overexpression of METTL3 elicited the opposite effects. Mechanistically, MeRIP-seq identified that METTL3 catalyzed m6A modification of TRAF1 mRNA and enhanced TRAF1 translation, thereby up-regulation of TRAF1 protein. Over-expression of TRAF1 successfully rescued the inhibition of proliferation and adhesion of endothelial cells due to METTL3 knockdown. Additionally, m6A methylation-mediated TRAF1 expression can be reversed by the demethylase ALKBH5. Knockdown of ALKBH5 upregulated the level of m6A and protein level of TRAF1, and also increased endothelial cells adhesion and inflammatory response. Collectively, our findings suggest that METTL3 regulates vascular endothelium homeostasis through TRAF1 m6A modification, suggesting that targeting the METTL3-m6A-TRAF1 axis may hold therapeutic potential for patients with vascular diseases.
Collapse
Affiliation(s)
- Duchu Chen
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.
| | - Wentao Xu
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Huaxian Zheng
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuxuan Zhang
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yongzhi Lin
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yulin Han
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Fenfen Yao
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Haohan Shen
- The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
26
|
Yan Y, Wang L, Zhong N, Wen D, Liu L. Multifaced roles of adipokines in endothelial cell function. Front Endocrinol (Lausanne) 2024; 15:1490143. [PMID: 39558976 PMCID: PMC11570283 DOI: 10.3389/fendo.2024.1490143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/14/2024] [Indexed: 11/20/2024] Open
Abstract
Obesity significantly contributes to the progression of cardiovascular diseases (CVDs) and elevates the risk of cardiovascular mortality. Atherosclerosis, the primary pathogenic process underlying CVDs, initiates with vascular endothelial dysfunction, serving as the cornerstone of vascular lesions. Adipokines, bioactive molecules secreted by adipose tissue that regulate metabolic and endocrine functions, play a pivotal role in modulating endothelial function during atherosclerosis. This review comprehensively examines the distinct roles of various adipokines in regulating endothelial function in atherosclerosis. We categorize these adipokines into two main groups: protective adipokines, including adiponectin, FGF21, CTRP9, PGRN, Omentin, and Vaspin, and detrimental adipokines such as leptin, Chemerin, Resistin, FABP4, among others. Targeting specific adipokines holds promise for novel clinical interventions in the management of atherosclerosis-related CVDs, thereby providing a theoretical foundation for cardiovascular disease treatment strategies.
Collapse
Affiliation(s)
- Yu Yan
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| | - Lihui Wang
- Department of Radiology, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Ni Zhong
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Donghua Wen
- Department of Laboratory Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Longhua Liu
- School of Exercise and Health, Shanghai University of Sport, Shanghai, China
| |
Collapse
|
27
|
Caliogna L, Berni M, Torriani C, Mancuso ME, Di Minno MND, Brancato AM, Jannelli E, Mosconi M, Pasta G. Pathogenesis of osteoarthritis, rheumatoid arthritis, and hemophilic arthropathy: The role of angiogenesis. Haemophilia 2024; 30:1256-1264. [PMID: 39297375 DOI: 10.1111/hae.15097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 09/06/2024] [Accepted: 09/06/2024] [Indexed: 12/21/2024]
Abstract
INTRODUCTION The term 'chronic inflammatory arthritis' (IA) can be used to define a group of heterogeneous diseases in which inflammation of the synovium is the common feature while having different pathogenesis and clinical outcomes. This condition can be found in osteoarthritis (OA), rheumatoid arthritis (RA), and hemophilic arthropathy (HA). AIM The objective is to try to highlight similarities and differences in the three pathological conditions and understand both molecular and physiological mechanisms. METHOD We have carried out a systematic review of the available literature following the guidelines Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA). RESULTS By comparing the data in the literature on OA, RA, and HA we have shown that the three pathologies differ in initial etiology but they motivate the same molecular pathways. CONCLUSION In this review we highlighted the similarities and differences between these diseases, creating ideas for future studies both in vivo and in vitro to develop new therapeutic agents and suggest possible biomarkers to follow the evolution and severity of the disease.
Collapse
Affiliation(s)
- Laura Caliogna
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Micaela Berni
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Camilla Torriani
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Maria Elisa Mancuso
- Center for Thrombosis and Hemorrhagic Diseases, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Alice Maria Brancato
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| | - Eugenio Jannelli
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Mario Mosconi
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
- Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, Pavia, Italy
| | - Gianluigi Pasta
- Orthopedics and Traumatology Clinic, IRCCS Policlinico San Matteo Foundation, Pavia, Italy
| |
Collapse
|
28
|
Smith EE, Biessels GJ, Gao V, Gottesman RF, Liesz A, Parikh NS, Iadecola C. Systemic determinants of brain health in ageing. Nat Rev Neurol 2024; 20:647-659. [PMID: 39375564 DOI: 10.1038/s41582-024-01016-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2024] [Indexed: 10/09/2024]
Abstract
Preservation of brain health is a worldwide priority. The traditional view is that the major threats to the ageing brain lie within the brain itself. Consequently, therapeutic approaches have focused on protecting the brain from these presumably intrinsic pathogenic processes. However, an increasing body of evidence has unveiled a previously under-recognized contribution of peripheral organs to brain dysfunction and damage. Thus, in addition to the well-known impact of diseases of the heart and endocrine glands on the brain, accumulating data suggest that dysfunction of other organs, such as gut, liver, kidney and lung, substantially affects the development and clinical manifestation of age-related brain pathologies. In this Review, a framework is provided to indicate how organ dysfunction can alter brain homeostasis and promote neurodegeneration, with a focus on dementia. We delineate the associations of subclinical dysfunction in specific organs with dementia risk and provide suggestions for public health promotion and clinical management.
Collapse
Affiliation(s)
- Eric E Smith
- Department of Clinical Neurosciences and Hotchkiss Brain Institute, University of Calgary, Calgary, Canada.
| | - Geert Jan Biessels
- Department of Neurology, UMC Utrecht Brain Center, University Medical Centre Utrecht, Utrecht, Netherlands
| | - Virginia Gao
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | | | - Arthur Liesz
- Institute for Stroke and Dementia Research, University Medical Center Munich, Munich, Germany
- Munich Cluster for Systems Neurology (SyNergy), Munich, Germany
| | - Neal S Parikh
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
29
|
Ruvira S, Rodríguez-Rodríguez P, Abderrahim F, Morales D, Cañas S, Valdivieso A, Ramiro-Cortijo D, Arribas SM. Resistance artery vasodilator pathways involved in the antihypertensive effects of cocoa shell extract in rats exposed to fetal undernutrition. J Physiol 2024; 602:6065-6085. [PMID: 39388282 DOI: 10.1113/jp287097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/20/2024] [Indexed: 10/12/2024] Open
Abstract
Fetal undernutrition establishes the foundations for hypertension development, with oxidative stress being a key hallmark. A growing interest in nutraceuticals for treating hypertension and environmental waste concerns prompted the present study aiming to evaluate whether supplementation with a polyphenol enriched extract from cocoa shell (CSE), a by-product from the chocolate industry with antioxidant properties, reduces hypertension of developmental origin, thus improving mesenteric resistance artery (MRA) vasodilatation. Adult male and female offspring from rats exposed to 50% food restriction from mid-gestation (maternal undernutrition, MUN) and controls were used. Supplementation was given through a gelatine (vehicle, VEH) or containing CSE (250 mg kg-1 day-1) 5 days week-1 for 3 weeks. Systolic blood pressure (SBP) was assessed by tail-cuff plethysmography. MRA function was studied by wire myography, and superoxide anion and nitric oxide were investigated by fluorescent indicators and confocal microscopy. Compared to control-VEH, MUN-VEH males showed significantly higher SBP, reduced MRA as well as relaxation to ACh, sodium nitroprusside and the AMPK agonist 5-aminoimidazole-4-carboxamide riboside, but not to isoproterenol. In MUN males, endothelial endothelium-derived hyperpolarizing factor and nitric oxide were unaltered, but MRA released a vasoconstrictor prostanoid and produced higher levels of superoxide anion. CSE normalized blood pressure and improved all above-mentioned MRA alterations in MUN males without an effect on control counterparts, except the reduction of superoxide anion. MUN-VEH females were normotensive and only showed a tendency towards larger superoxide anion production, which was abolished by CSE. CSE supplementation reduces SBP improving endothelium-dependent and independent MRA vasodilatation, related to local superoxide anion reduction, being a potential nutraceutical ingredient to counteract hypertension, in addition to contributing to the circular economy. KEY POINTS: Fetal undernutrition induces hypertension in males associated with deficient resistance artery vasodilatation, being normalized by cocoa shell extract (CSE). Release of a cyclooxygenase-derived contractile factor is the main endothelial alteration, which is abolished by CSE. AMPK and soluble guanylyl cyclase-mediated relaxation are also reduced in smooth muscle cells from maternal undernutrition resistance arteries, being improved by CSE. Vascular oxidative damage caused by excess superoxide anion generation can account for impaired vasodilatation, which is improved by CSE. The capacity of CSE to improve relaxation is probably related to its antioxidant bioactive factors, and thus cocoa shell is a potential food by-product to treat hypertension.
Collapse
Affiliation(s)
- Santiago Ruvira
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pilar Rodríguez-Rodríguez
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| | - Fatima Abderrahim
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - Dolores Morales
- Confocal Microscopy Service (SiDI), Faculty of Medicine, Universidad Autonoma de Madrid, Madrid, Spain
| | - Silvia Cañas
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
- Institute of Food Science Research (CIAL), Universidad Autónoma de Madrid (UAM-CSIC), Madrid, Spain
- Department of Agricultural Chemistry and Food Science, Faculty of Science, Universidad Autónoma de Madrid, Madrid, Spain
| | - Alberto Valdivieso
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - David Ramiro-Cortijo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| | - Silvia M Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Food, Oxidative Stress and Cardiovascular Health (FOSCH) Research Group, Universidad Autónoma de Madrid, Madrid, Spain
| |
Collapse
|
30
|
Daniele A, Lucas SJE, Rendeiro C. Variability of flow-mediated dilation across lower and upper limb conduit arteries. Eur J Appl Physiol 2024; 124:3265-3278. [PMID: 38878074 PMCID: PMC11519148 DOI: 10.1007/s00421-024-05517-z] [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: 11/06/2023] [Accepted: 05/23/2024] [Indexed: 10/30/2024]
Abstract
Endothelial dysfunction is an early predictor of atherosclerosis and cardiovascular disease. Flow-mediated dilation (FMD) is the gold standard to assess endothelial function in humans. FMD reproducibility has been mainly assessed in the brachial artery (BA) with limited research in lower limb arteries. The purpose of this study was to compare FMD reproducibility in the upper limb BA and lower limb superficial femoral artery (SFA) in young healthy adults.Fifteen young healthy adults (nine males; six females) underwent FMD, resting diameter, velocity, and shear rate measurements on three occasions to determine intra-and inter-day reproducibility in both BA and SFA, assessed by coefficient of variation (CV), intraclass correlation coefficient (ICC), and Bland-Altman plots.BA FMD CVs (intra-day: 4.2%; inter-day: 8.7%) and ICCs (intra-day: 0.967; inter-day: 0.903) indicated excellent reproducibility and reliability, while for SFA FMD, both CVs (intra-day: 11.6%; inter-day: 26.7%) and ICCs (intra-day: 0.898; inter-day: 0.651) showed good/moderate reproducibility and reliability. BA FMD was significantly more reproducible than SFA FMD (p < 0.05). Diameter reproducibility was excellent and similar between arteries, while resting velocity and shear rate have lower reproducibility in the BA compared to SFA. Bland-Altman plots displayed no proportional and fixed bias between measurements.In summary, SFA FMD is less reproducible than BA FMD, with identical volume of ultrasound training. Given the increasing interest in using SFA FMD to test the efficacy of interventions targeting lower limb's vascular health and as a potential biomarker for peripheral arterial disease risk, future studies should ensure higher levels of training for adequate reproducibility.
Collapse
Affiliation(s)
- Alessio Daniele
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
| | - Samuel J E Lucas
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK
| | - Catarina Rendeiro
- School of Sport, Exercise and Rehabilitation Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
- Centre for Human Brain Health, University of Birmingham, Birmingham, UK.
| |
Collapse
|
31
|
Ju NR, Fakhr MS, Gholami P, Khosravi F, Rezvanfar K. Refractory chylothorax following COVID-19: Successful surgical management of a rare complication; a case report. Int J Surg Case Rep 2024; 124:110403. [PMID: 39366117 PMCID: PMC11483476 DOI: 10.1016/j.ijscr.2024.110403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/30/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024] Open
Abstract
INTRODUCTION AND IMPORTANCE Chylothorax is an uncommon complication linked to COVID-19. The fundamental pathophysiology and most effective management strategy are still uncertain. CASE PRESENTATION A 72-year-old man presented with worsening dyspnea and fatigue one month post-COVID-19. Imaging demonstrated a significant right pleural effusion, with thoracentesis confirming the presence of chylothorax. Despite the implementation of conservative interventions, the effusion remained unresolved. During the right thoracotomy procedure, a 1 cm (about 0.39 in) perforation in proximity to the Azygos vein, encircled by hypertrophic lymph nodes, was identified. Surgical intervention successfully alleviated the symptoms. CLINICAL DISCUSSION This case implies that mediastinal lymphadenopathy because of COVID-19 could potentially obstruct and interfere with the thoracic duct. This emphasizes the significance of considering chylothorax as a crucial diagnostic possibility in individuals presenting with new onset pleural effusions following COVID-19. Although conservative approaches are typically the first line of management, persistent cases may necessitate surgical intervention to target the root cause. CONCLUSIONS Additional research is imperative to elucidate the intricate pathways connecting COVID-19 and chylothorax, as well as to ascertain the most effective diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Nasrin Rahmani Ju
- Department of Surgery, Farhikhtegan Research Center, Tehran Medical Sciences School Branch, Islamic Azad University, Tehran, Iran
| | - Masoud Saadat Fakhr
- Faculty of Medicine, Tehran Medical Sciences School Branch, Islamic Azad University, Tehran, Iran
| | - Poorya Gholami
- Faculty of Medicine, Tehran Medical Sciences School Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Khosravi
- Faculty of Medicine, Tehran Medical Sciences School Branch, Islamic Azad University, Tehran, Iran
| | - Kiana Rezvanfar
- Faculty of Medicine, Tehran Medical Sciences School Branch, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
32
|
Zhang X, Yang J, Lu Y, Liu Y, Wang T, Yu F. Human Urinary Kallidinogenase improves vascular endothelial injury by activating the Nrf2/HO-1 signaling pathway. Chem Biol Interact 2024; 403:111230. [PMID: 39244186 DOI: 10.1016/j.cbi.2024.111230] [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: 06/06/2024] [Revised: 08/16/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Vascular endothelial injury is closely related to the progression of various cardio-cerebrovascular diseases. Whether Human Urinary Kallidinogenase (HUK) has a protective effect on endothelial injury remains unclear. This study established an in vivo model of rat common carotid artery intima injury and an in vitro model of human umbilical vein endothelial cell (HUVECs) injury induced by hydrogen peroxide (H2O2). To explore the protective effect and mechanism of HUK on endothelial injury. In vivo, HUK can reduce the hyperplasia and lumen stenosis of rat common carotid artery after intimal injury, and promote the fluorescence expression of vWF in the common carotid artery. HUK also activated the Nrf2/HO-1 signaling pathway in rat common carotid artery tissue to reduce endothelial damage. In vitro, HUK can inhibit the H2O2-induced decline in HUVECs activity, improve the migration ability of HUVECs induced by H2O2, inhibit the apoptosis and necrosis of HUVECs and the generation of ROS, and regulate the expression of VEGFA, ET-1 and eNOS proteins related to endothelial function in cells. The Nrf2/HO-1 signaling pathway is activated, and the HO-1 specific inhibitor zinc porphyrin (ZnPP) can partially reverse the protective effect of HUK on H2O2-induced HUVECs injury in terms of cell migration, necrosis and oxidative stress. The Nrf2/HO-1 signaling pathway plays an important role in the regulation of migration, necrosis and oxidative stress of HUVECs cells. HUK has a protective effect on vascular endothelial injury. HUK can inhibit oxidative stress and apoptotic necrosis by activating Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Xiong Zhang
- Department of Clinical Pharmacology Lab, Nanjing First Hospital, Nanjing Medical University, Jiangsu, 210001, PR China
| | - Jiaying Yang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, 211100, PR China
| | - Yini Lu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, 211100, PR China
| | - Yi Liu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, 211100, PR China
| | - Tianyin Wang
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, 211100, PR China
| | - Feng Yu
- Department of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Jiangsu, 211100, PR China.
| |
Collapse
|
33
|
Wang X, He M, Yu R, Tan L. Porous boron-doped graphitic carbon nitride-based label-free electrochemical immunoassay of vascular endothelial cadherin. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:7114-7120. [PMID: 39308352 DOI: 10.1039/d4ay01112k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
As a main connecting protein between endothelial cells, vascular endothelial cadherin (CD144) is involved in regulating vascular remodeling and maintaining vascular integrity. It is regarded as a marker of endothelial dysfunction and injury. Quantitative determination of CD144 is of importance in pathology research, diagnosis and treatment of vascular diseases. A label-free electrochemical method for the immunoassay of CD144 was developed in this work. CD144 antibodies were assembled on a glassy carbon electrode modified with porous boron-doped carbon nitride (B-GCN) and gold nanoparticles (AuNPs) in the presence of protein A. The binding of CD144 on the antibody-modified electrode induced serious steric hindrance, inhibiting the diffusion of ferri-/ferrocyanide from the bulk electrolyte to the electrode interface. The change of the differential pulse voltammetric response displayed a linear relationship with the concentration of CD144 between 0.500 and 400 ng mL-1. The new electrochemical sensor showed some good performances including good selectivity, high stability and satisfactory reproducibility. The cellular morphology observation and activity measurement showed that the dysfunction of vascular endothelial cells appeared in the presence of high-content NaCl. The electrochemical analysis reveals a positive correlation between the release amount of CD144 from the dysfunctional cells and the NaCl concentration in the growth medium.
Collapse
Affiliation(s)
- Xuan Wang
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| | - Mengting He
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| | - Rujie Yu
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| | - Liang Tan
- College of Chemistry and Chemical Engineering, Hunan Normal University, Changsha, 410081, PR China.
| |
Collapse
|
34
|
Ma Y, Song D, Yuan J, Hao W, Xi J, Yuan C, Cheng Z. Alisol A inhibits and stabilizes atherosclerotic plaques by protecting vascular endothelial cells. Front Pharmacol 2024; 15:1493948. [PMID: 39525632 PMCID: PMC11543447 DOI: 10.3389/fphar.2024.1493948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 09/30/2024] [Indexed: 11/16/2024] Open
Abstract
Background and aims Dysfunction of endothelial cells represents a crucial aspect in the pathogenesis of atherosclerosis. The aim of this study was to explore the protective effects of alisol A on vascular endothelial cells and its possible mechanisms. Methods An atherosclerosis model was established by feeding ApoE-/- mice with high-fat chow. Alisol A (150 mg/kg/d) or atorvastatin (15 mg/kg/d) was administered, and the levels of blood lipids were evaluated. The effect of the drugs on atherosclerotic plaques was observed by staining the aorta with Sudan IV. In vitro experiments were conducted using human aortic endothelial cells (HAECs) to assess the effects of alisol A on cell proliferation, migration, tubulation, secretion, and cellular integrity by CCK-8 assay, wound healing assay, angiogenesis assay, NO secretion, and release of LDH. Transcriptomics and molecular docking were used to explore the mechanism of plaque inhibition and stabilization by alisol A. Results Alisol A significantly reduced the aortic plaque area in ApoE-/- mice fed with high-fat chow. In vitro, alisol A had a protective effect on HAECs, which was reflected in the inhibition of vascular endothelial cell proliferation, promotion of NO secretion by vascular endothelial cells, inhibition of vascular endothelial cell migration and angiogenesis, and the maintenance of cell membrane integrity. Therefore, alisol A inhibited and stabilized atherosclerotic plaques and slowed down the process of atherosclerosis. Transcriptomics studies showed 4,086 differentially expressed genes (DEGs) in vascular endothelial cells after alisol A treatment. Enrichment analysis indicated that many genes involved in TNF signaling pathway were differentially expressed, and inflammatory genes were suppressed. The molecular docking results verified the hypothesis that alisol A has a low binding energy after docking with TNF target, and TNF could be a potential target of alisol A. Conclusion Alisol A produced protection on vascular endothelial cells, achieving inhibition and stabilization of atherosclerotic plaques.
Collapse
Affiliation(s)
- Yang Ma
- China State Institute of Pharmaceutical Industry, National Advanced Medical Engineering Research Center, Shanghai, China
| | - Dingzhong Song
- China State Institute of Pharmaceutical Industry, National Advanced Medical Engineering Research Center, Shanghai, China
| | - Jie Yuan
- China State Institute of Pharmaceutical Industry, National Advanced Medical Engineering Research Center, Shanghai, China
| | - Wusi Hao
- China State Institute of Pharmaceutical Industry, National Advanced Medical Engineering Research Center, Shanghai, China
| | - Jianqiang Xi
- China State Institute of Pharmaceutical Industry, National Advanced Medical Engineering Research Center, Shanghai, China
| | - Chunping Yuan
- Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai, China
| | - Zhihong Cheng
- China State Institute of Pharmaceutical Industry, National Advanced Medical Engineering Research Center, Shanghai, China
| |
Collapse
|
35
|
Wang X, Yang X, Qi X, Fan G, Zhou L, Peng Z, Yang J. Anti-atherosclerotic effect of incretin receptor agonists. Front Endocrinol (Lausanne) 2024; 15:1463547. [PMID: 39493783 PMCID: PMC11527663 DOI: 10.3389/fendo.2024.1463547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 09/30/2024] [Indexed: 11/05/2024] Open
Abstract
Incretin receptor agonists (IRAs), primarily composed of glucagon-like peptide-1 receptor agonists (GLP-1RAs) and glucose-dependent insulinotropic polypeptide receptor agonists (GIPRAs), work by mimicking the actions of the endogenous incretin hormones in the body. GLP-1RAs have been approved for use as monotherapy and in combination with GIPRAs for the management of type 2 diabetes mellitus (T2DM). In addition to their role in glucose regulation, IRAs have demonstrated various benefits such as cardiovascular protection, obesity management, and regulation of bone turnover. Some studies have suggested that IRAs not only aid in glycemic control but also exhibit anti-atherosclerotic effects. These agents have been shown to modulate lipid abnormalities, reduce blood pressure, and preserve the structural and functional integrity of the endothelium. Furthermore, IRAs have the ability to mitigate inflammation by inhibiting macrophage activation and promoting M2 polarization. Research has also indicated that IRAs can decrease macrophage foam cell formation and prevent vascular smooth muscle cell (VSMC) phenotype switching, which are pivotal in atheromatous plaque formation and stability. This review offers a comprehensive overview of the protective effects of IRAs in atherosclerotic disease, with a focus on their impact on atherogenesis.
Collapse
Affiliation(s)
- Xin Wang
- Department of Metabolism and Endocrinology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin Yang
- Department of Metabolism and Endocrinology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaoyan Qi
- Department of Metabolism and Endocrinology, Shenzhen Nanshan People's Hospital; The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Gang Fan
- Department of Urology, Shenzhen Nanshan People's Hospital; The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Lingzhi Zhou
- Department of pediatrics, Shenzhen Nanshan People's Hospital; The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| | - Zhengliang Peng
- Department of Emergency, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Jing Yang
- Department of Metabolism and Endocrinology, the First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Metabolism and Endocrinology, Shenzhen Nanshan People's Hospital; The Sixth Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, Guangdong, China
| |
Collapse
|
36
|
Vakili S, Izydore EK, Losert L, Cabral WA, Tavarez UL, Shores K, Xue H, Erdos MR, Truskey GA, Collins FS, Cao K. Angiopoietin-2 reverses endothelial cell dysfunction in progeria vasculature. Aging Cell 2024:e14375. [PMID: 39422121 DOI: 10.1111/acel.14375] [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: 05/09/2024] [Revised: 08/31/2024] [Accepted: 09/13/2024] [Indexed: 10/19/2024] Open
Abstract
Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder in children caused by a point mutation in the lamin A gene, resulting in a toxic form of lamin A called progerin. Accelerated atherosclerosis leading to heart attack and stroke are the major causes of death in these patients. Endothelial cell (EC) dysfunction contributes to the pathogenesis of HGPS related cardiovascular diseases (CVD). Endothelial cell-cell communications are important in the development of the vasculature, and their disruptions contribute to cardiovascular pathology. However, it is unclear how progerin interferes with such communications that lead to vascular dysfunction. An antibody array screening of healthy and HGPS patient EC secretomes identified Angiopoietin-2 (Ang2) as a down-regulated signaling molecule in HGPS ECs. A similar down-regulation of Ang2 mRNA and protein was detected in the aortas from an HGPS mouse model. Addition of Ang2 to HGPS ECs rescues vasculogenesis, normalizes endothelial cell migration and gene expression, and restores nitric oxide bioavailability through eNOS activation. Furthermore, Ang2 addition reverses unfavorable paracrine effects of HGPS ECs on vascular smooth muscle cells. Lastly, by utilizing adenine base editor (ABE)-corrected HGPS ECs and progerin-expressing HUVECs, we demonstrated a negative correlation between progerin and Ang2 expression. Lastly, our results indicated that Ang2 exerts its beneficial effect in ECs through Tie2 receptor binding, activating an Akt-mediated pathway. Together, these results provide molecular insights into EC dysfunction in HGPS and suggest that Ang2 treatment has potential therapeutic effects in HGPS-related CVD.
Collapse
Affiliation(s)
- Sahar Vakili
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Elizabeth K Izydore
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Leonhard Losert
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| | - Wayne A Cabral
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Urraca L Tavarez
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kevin Shores
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Huijing Xue
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
- Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Michael R Erdos
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - George A Truskey
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Francis S Collins
- Molecular Genetics Section, Center for Precision Health Research, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kan Cao
- Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, Maryland, USA
| |
Collapse
|
37
|
Passier M, Bentley K, Loerakker S, Ristori T. YAP/TAZ drives Notch and angiogenesis mechanoregulation in silico. NPJ Syst Biol Appl 2024; 10:116. [PMID: 39368976 PMCID: PMC11455968 DOI: 10.1038/s41540-024-00444-3] [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: 04/30/2024] [Accepted: 09/22/2024] [Indexed: 10/07/2024] Open
Abstract
Endothelial cells are key players in the cardiovascular system. Among other things, they are responsible for sprouting angiogenesis, the process of new blood vessel formation essential for both health and disease. Endothelial cells are strongly regulated by the juxtacrine signaling pathway Notch. Recent studies have shown that both Notch and angiogenesis are influenced by extracellular matrix stiffness; however, the underlying mechanisms are poorly understood. Here, we addressed this challenge by combining computational models of Notch signaling and YAP/TAZ, stiffness- and cytoskeleton-regulated mechanotransducers whose activity inhibits both Dll4 (Notch ligand) and LFng (Notch-Dll4 binding modulator). Our simulations successfully mimicked previous experiments, indicating that this YAP/TAZ-Notch crosstalk elucidates the Notch and angiogenesis mechanoresponse to stiffness. Additional simulations also identified possible strategies to control Notch activity and sprouting angiogenesis via cytoskeletal manipulations or spatial patterns of alternating stiffnesses. Our study thus inspires new experimental avenues and provides a promising modeling framework for further investigations into the role of Notch, YAP/TAZ, and mechanics in determining endothelial cell behavior during angiogenesis and similar processes.
Collapse
Affiliation(s)
- Margot Passier
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Katie Bentley
- The Francis Crick Institute, London, UK
- Department of Informatics, King's College London, London, UK
| | - Sandra Loerakker
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Tommaso Ristori
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands.
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands.
| |
Collapse
|
38
|
Guelfi S, Hodivala-Dilke K, Bergers G. Targeting the tumour vasculature: from vessel destruction to promotion. Nat Rev Cancer 2024; 24:655-675. [PMID: 39210063 DOI: 10.1038/s41568-024-00736-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
As angiogenesis was recognized as a core hallmark of cancer growth and survival, several strategies have been implemented to target the tumour vasculature. Yet to date, attempts have rarely been so diverse, ranging from vessel growth inhibition and destruction to vessel normalization, reprogramming and vessel growth promotion. Some of these strategies, combined with standard of care, have translated into improved cancer therapies, but their successes are constrained to certain cancer types. This Review provides an overview of these vascular targeting approaches and puts them into context based on our subsequent improved understanding of the tumour vasculature as an integral part of the tumour microenvironment with which it is functionally interlinked. This new knowledge has already led to dual targeting of the vascular and immune cell compartments and sets the scene for future investigations of possible alternative approaches that consider the vascular link with other tumour microenvironment components for improved cancer therapy.
Collapse
Affiliation(s)
- Sophie Guelfi
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium
| | - Kairbaan Hodivala-Dilke
- Barts Cancer Institute, Queen Mary University of London, John Vane Science Centre, London, UK.
| | - Gabriele Bergers
- Department of Oncology, VIB-KU Leuven Center for Cancer Biology and KU Leuven, Leuven, Belgium.
| |
Collapse
|
39
|
Seo BG, Lee IW, Kim HJ, Lee YJ, Kim O, Lee JH, Lee JH, Hwangbo C. Angiogenic properties and intercellular communication of differentiated porcine endothelial cells in vascular therapy. Sci Rep 2024; 14:22844. [PMID: 39354086 PMCID: PMC11445381 DOI: 10.1038/s41598-024-73584-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 09/18/2024] [Indexed: 10/03/2024] Open
Abstract
Endothelial cell dysfunction can lead to various vascular diseases. Blood flow disorder is a common symptom of vascular diseases. Regenerative angiogenesis, which involves transplanting vascular cells or stem cells into the body to shape new vasculature, can be a good therapeutic strategy. However, there are several limitations to using autologous cells from the patients themselves. We sought to investigate the new vascular cells that can play a role in the formation of angiogenesis in vivo using stem cells from alternative animals suitable for cellular therapy. Porcine is an optimal animal model for xenotransplantation owing to its physiological similarity to humans. We used differentiated porcine endothelial cells (pECs) as a therapeutic strategy to restore vessel function. Differentiated pECs formed vessel-like structures in mice, distinguishing them from stem cells. MMPs activity and migration assays indicated that differentiated pECs possessed angiogenic potential. Tube formation and 3D spheroid sprouting assays further confirmed the angiogenic phenotype of the differentiated pECs. Immunofluorescence and immunoprecipitation analyses revealed claudin-mediated tight junctions and connexin 43-mediated gap junctions between human ECs and differentiated pECs. Additionally, the movement of small RNA from human ECs to differentiated pECs was observed under co-culture conditions. Our findings demonstrated the in vivo viability and angiogenetic potential of differentiated pECs and highlighted the potential for intercellular communication between human and porcine ECs. These results suggest that transplanted cells in vascular regeneration completed after cell therapy have the potential to achieve intercellular communication within the body.
Collapse
Affiliation(s)
- Bo-Gyeong Seo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - In-Won Lee
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
- Department of Animal Bioscience, College of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Hyo-Jin Kim
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Yeon-Ji Lee
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
- Department of Animal Bioscience, College of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea
| | - Okhwa Kim
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea
| | - Joon-Hee Lee
- Department of Animal Bioscience, College of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
- Institute of Agriculture and Life Science, College of Agriculture & Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| | - Jeong-Hyung Lee
- Kangwon Institute of Inclusive Technology, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea.
- Department of Biochemistry (BK21 Four), College of Natural Sciences, Kangwon National University, Chuncheon, Gangwon-do, 24341, Republic of Korea.
| | - Cheol Hwangbo
- Division of Life Science, College of Natural Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
- Division of Applied Life Science (BK21 Four), Research Institute of Life Sciences, Gyeongsang National University, Jinju, 52828, Republic of Korea.
| |
Collapse
|
40
|
Luo Z, Tang YY, Zhou L. Melatonin as an adjunctive therapy in cardiovascular disease management. Sci Prog 2024; 107:368504241299993. [PMID: 39574322 PMCID: PMC11585022 DOI: 10.1177/00368504241299993] [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] [Indexed: 11/25/2024]
Abstract
Melatonin, N-acetyl-5-methoxytryptamine, is a neuroendocrine hormone secreted by the pineal gland. This pleiotropic indoleamine possesses amphiphilic properties, allowing it to penetrate most biological barriers and exert its effects at the subcellular level. Importantly, melatonin also plays a crucial role in regulating the body's response to circadian rhythms, adapting to internal and external environmental cues. Melatonin functions as a powerful antioxidant and free radical scavenger, protecting cells from oxidative damage. Its diverse physiological roles include maintaining the functional integrity of endothelial cells, thereby preventing atherosclerosis, a major contributor to cardiovascular disease. Additionally, melatonin exhibits antioxidant and free radical scavenging properties, potentially improving metabolic disorders. These combined effects suggest a unique adjunctive therapeutic potential for melatonin in treating cardiovascular diseases. This review aims to explore the mechanisms by which melatonin interacts with the cardiovascular system and investigates its potential use as an adjunctive therapeutic agent in managing cardiovascular disease.
Collapse
Affiliation(s)
- Zan Luo
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuan Yuan Tang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Liang Zhou
- Department of Cardiovascular Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Clinical Medicine, Hangzhou, China
| |
Collapse
|
41
|
Craige SM, Kaur G, Bond JM, Caliz AD, Kant S, Keaney JF. Endothelial Reactive Oxygen Species: Key Players in Cardiovascular Health and Disease. Antioxid Redox Signal 2024. [PMID: 39213161 DOI: 10.1089/ars.2024.0706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Significance: Endothelial cells (ECs) line the entire vasculature system and serve as both barriers and facilitators of intra- and interorgan communication. Positioned to rapidly sense internal and external stressors, ECs dynamically adjust their functionality. Endothelial dysfunction occurs when the ability of ECs to react to stressors is impaired, which precedes many cardiovascular diseases (CVDs). While EC reactive oxygen species (ROS) have historically been implicated as mediators of endothelial dysfunction, more recent studies highlight the central role of ROS in physiological endothelial signaling. Recent Advances: New evidence has uncovered that EC ROS are fundamental in determining how ECs interact with their environment and respond to stress. EC ROS levels are mediated by external factors such as diet and pathogens, as well as inherent characteristics, including sex and location. Changes in EC ROS impact EC function, leading to changes in metabolism, cell communication, and potentially disrupted signaling in CVDs. Critical Issues: Current endothelial biology concepts integrate the dual nature of ROS, emphasizing the importance of EC ROS in physiological stress adaptation and their contribution to CVDs. Understanding the discrete, localized signaling of EC ROS will be critical in preventing adverse cardiovascular outcomes. Future Directions: Exploring how the EC ROS environment alters EC function and cross-cellular communication is critical. Considering the inherent heterogeneity among EC populations and understanding how EC ROS contribute to this diversity and the role of sexual dimorphism in the EC ROS environment will be fundamental for developing new effective cardiovascular treatment strategies.
Collapse
Affiliation(s)
- Siobhan M Craige
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, USA
| | - Gaganpreet Kaur
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Jacob M Bond
- Department of Human Nutrition, Foods, and Exercise, Virginia Tech, Blacksburg, Virginia, USA
- Translational Biology, Medicine, and Health Program, Virginia Tech, Roanoke, Virginia, USA
| | - Amada D Caliz
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Shashi Kant
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - John F Keaney
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
42
|
Tiwari P, Elgazzaz M, Lazartigues E, Hanif K. Effect of Diminazene Aceturate, an ACE2 activator, on platelet CD40L signaling induced glial activation in rat model of hypertension. Int Immunopharmacol 2024; 139:112654. [PMID: 38996777 DOI: 10.1016/j.intimp.2024.112654] [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: 10/24/2023] [Revised: 06/30/2024] [Accepted: 07/06/2024] [Indexed: 07/14/2024]
Abstract
Hypertension causes platelet activation and adhesion in the brain resulting in glial activation and neuroinflammation. Further, activation of Angiotensin-Converting Enzyme 2/Angiotensin (1-7)/Mas Receptor (ACE2/Ang (1-7)/MasR) axis of central Renin-Angiotensin System (RAS), is known to reduce glial activation and neuroinflammation, thereby exhibiting anti-hypertensive and anti-neuroinflammatory properties. Therefore, in the present study, the role of ACE2/Ang (1-7)/MasR axis was studied on platelet-induced glial activation and neuroinflammation using Diminazene Aceturate (DIZE), an ACE2 activator, in astrocytes and microglial cells as well as in rat model of hypertension. We found that the ACE2 activator DIZE, independently of its BP-lowering properties, efficiently prevented hypertension-induced glial activation, neuroinflammation, and platelet CD40-CD40L signaling via upregulation of ACE2/Ang (1-7)/MasR axis. Further, DIZE decreased platelet deposition in the brain by reducing the expression of adhesion molecules on the brain endothelium. Activation of ACE2 also reduced hypertension-induced endothelial dysfunction by increasing eNOS bioavailability. Interestingly, platelets isolated from hypertensive rats or activated with ADP had significantly increased sCD40L levels and induced significantly more glial activation than platelets from DIZE treated group. Therefore, injection of DIZE pre-treated ADP-activated platelets into normotensive rats strongly reduced glial activation compared to ADP-treated platelets. Moreover, CD40L-induced glial activation, CD40 expression, and NFкB-NLRP3 inflammatory signaling are reversed by DIZE. Furthermore, the beneficial effects of ACE2 activation, DIZE was found to be significantly blocked by MLN4760 (ACE2 inhibitor) as well as A779 (MasR antagonist) treatments. Hence, our study demonstrated that ACE2 activation reduced the platelet CD40-CD40L induced glial activation and neuroinflammation, hence imparted neuroprotection.
Collapse
Affiliation(s)
- Priya Tiwari
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Mona Elgazzaz
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Eric Lazartigues
- Department of Pharmacology & Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Kashif Hanif
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow 226031, U.P., India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
43
|
Ryu JY, Zhang J, Tirado SR, Dagen S, Frerichs KU, Patel NJ, Aziz-Sultan MA, Brown A, Rogers-Grazado M, Amr SS, Weiss ST, Du R. MiRNA expression profiling reveals a potential role of microRNA-148b-3p in cerebral vasospasm in subarachnoid hemorrhage. Sci Rep 2024; 14:22539. [PMID: 39341923 PMCID: PMC11438990 DOI: 10.1038/s41598-024-73579-2] [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: 03/20/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Cerebral vasospasm (CVS) is an important contributor to delayed cerebral ischemia following aneurysmal subarachnoid hemorrhage (aSAH), leading to high morbidity and long-term disability. While several microRNAs (miRNAs) have been implicated in vasospasm, the underlying mechanisms for CVS remain poorly understood. Our study aims to identify miRNAs that may contribute to the development of CVS. Whole-blood samples were obtained during or outside of vasospasm from aSAH patients whose maximal vasospasm was moderate or severe. MiRNAs were isolated from serial whole-blood samples, and miRNA sequencing was performed. Differentially expressed miRNAs were identified and the expression levels in patients' samples were verified using real-time qPCR. The biological functions of identified miRNA were evaluated in human brain endothelial cells (HBECs). MiRNA profiling revealed significant upregulation of miR-148b-3p in patients during CVS. We demonstrated that miR-148b-3p directly targeted and decreased the expression of ROCK1, affecting cell proliferation, migration, and invasion of HBECs through the ROCK-LIMK-Cofilin pathway. We propose that the upregulation of miRNA-148b-3p plays a role in the development of CVS by regulating actin cytoskeletal dynamics in HBECs, which is crucial for vascular function. Our study highlights miR-148b-3p as a potential diagnostic marker as well as therapeutic target for CVS following aSAH.
Collapse
Affiliation(s)
- Jee-Yeon Ryu
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Jianing Zhang
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Selena-Rae Tirado
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Sarajune Dagen
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Kai U Frerichs
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Nirav J Patel
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - M Ali Aziz-Sultan
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA
| | - Alison Brown
- Mass General Brigham Personalized Medicine, Mass General Brigham, Cambridge, MA, USA
| | | | - Sami S Amr
- Mass General Brigham Personalized Medicine, Mass General Brigham, Cambridge, MA, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Rose Du
- Department of Neurosurgery, Brigham and Women's Hospital, Harvard Medical School, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
44
|
de Lima EP, Tanaka M, Lamas CB, Quesada K, Detregiachi CRP, Araújo AC, Guiguer EL, Catharin VMCS, de Castro MVM, Junior EB, Bechara MD, Ferraz BFR, Catharin VCS, Laurindo LF, Barbalho SM. Vascular Impairment, Muscle Atrophy, and Cognitive Decline: Critical Age-Related Conditions. Biomedicines 2024; 12:2096. [PMID: 39335609 PMCID: PMC11428869 DOI: 10.3390/biomedicines12092096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/22/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
The triad of vascular impairment, muscle atrophy, and cognitive decline represents critical age-related conditions that significantly impact health. Vascular impairment disrupts blood flow, precipitating the muscle mass reduction seen in sarcopenia and the decline in neuronal function characteristic of neurodegeneration. Our limited understanding of the intricate relationships within this triad hinders accurate diagnosis and effective treatment strategies. This review analyzes the interrelated mechanisms that contribute to these conditions, with a specific focus on oxidative stress, chronic inflammation, and impaired nutrient delivery. The aim is to understand the common pathways involved and to suggest comprehensive therapeutic approaches. Vascular dysfunctions hinder the circulation of blood and the transportation of nutrients, resulting in sarcopenia characterized by muscle atrophy and weakness. Vascular dysfunction and sarcopenia have a negative impact on physical function and quality of life. Neurodegenerative diseases exhibit comparable pathophysiological mechanisms that affect cognitive and motor functions. Preventive and therapeutic approaches encompass lifestyle adjustments, addressing oxidative stress, inflammation, and integrated therapies that focus on improving vascular and muscular well-being. Better understanding of these links can refine therapeutic strategies and yield better patient outcomes. This study emphasizes the complex interplay between vascular dysfunction, muscle degeneration, and cognitive decline, highlighting the necessity for multidisciplinary treatment approaches. Advances in this domain promise improved diagnostic accuracy, more effective therapeutic options, and enhanced preventive measures, all contributing to a higher quality of life for the elderly population.
Collapse
Affiliation(s)
- Enzo Pereira de Lima
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
| | - Masaru Tanaka
- HUN-REN-SZTE Neuroscience Research Group, Danube Neuroscience Research Laboratory, Hungarian Research Network, University of Szeged (HUN-REN-SZTE), Tisza Lajos Krt. 113, H-6725 Szeged, Hungary
| | - Caroline Barbalho Lamas
- Department of Gerontology, Universidade Federal de São Carlos, UFSCar, São Carlos 13565-905, SP, Brazil
| | - Karina Quesada
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
| | - Claudia Rucco P. Detregiachi
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
| | - Adriano Cressoni Araújo
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Elen Landgraf Guiguer
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Virgínia Maria Cavallari Strozze Catharin
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Marcela Vialogo Marques de Castro
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Department of Odontology, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Edgar Baldi Junior
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Marcelo Dib Bechara
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | | | | | - Lucas Fornari Laurindo
- Department of Biochemistry and Pharmacology, School of Medicine, Faculdade de Medicina de Marília (FAMEMA), Marília 17525-902, SP, Brazil
- Department of Administration, Associate Degree in Hospital Management, Universidade de Marília (UNIMAR), Marília 17525-902, SP, Brazil
| | - Sandra Maria Barbalho
- Department of Biochemistry and Pharmacology, School of Medicine, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil (M.D.B.)
- Postgraduate Program in Structural and Functional Interactions in Rehabilitation, University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
- Research Coordination, UNIMAR Charity Hospital (HBU), University of Marília (UNIMAR), Marília 17525-902, SP, Brazil
| |
Collapse
|
45
|
Sarkar P, Amin KN, Balakrishnan R, Ramkumar KM, Arockiaraj J. Molecular Mechanism of NL13 Peptide of Adenosyl Homocysteinase Against ER Stress through Nrf2 Signaling Cascade. Int J Pept Res Ther 2024; 30:58. [DOI: 10.1007/s10989-024-10637-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2024] [Indexed: 01/12/2025]
|
46
|
Desanlis J, Gordon D, French C, Calveyrac C, Cottin F, Gernigon M. Effects of occlusion pressure on hemodynamic responses recorded by near-infrared spectroscopy across two visits. Front Physiol 2024; 15:1441239. [PMID: 39324105 PMCID: PMC11422206 DOI: 10.3389/fphys.2024.1441239] [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: 05/30/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Ischemic Preconditioning (IPC) has emerged as a promising approach to mitigate the impact of hypoxia on physiological functions. However, the heterogeneity of occlusion pressures for inducing arterial occlusion has led to inconsistent hemodynamic outcomes across studies. This study aims to evaluate the peripheral hemodynamic responses to partial and total blood-flow occlusions on the left arm at rest, using absolute or individualized pressures, on two occasions. Thirty-five young males volunteered to participate in this study. IPC procedure (3 × 7-min) was performed on the left upper arm with cuff pressures at 50 mmHg (G1), 50 mmHg over the systolic blood pressure (SBP + 50 mmHg) (G2) or 250 mmHg (G3). NIRS-derived parameters were assessed for each occlusion and reperfusion phase in the brachioradialis. Results showed a significantly lower magnitude of deoxygenation (TSIAUC) for G1 compared to G2 (-1959.2 ± 1417.4 vs. -10908.1 ± 1607.5, P < 0.001) and G3 -1959.2 ± 1417.4 vs. -11079.3 ± 1828.1, P < 0.001), without differences between G2 and G3. However, G3 showed a significantly faster reoxygenation only for tissue saturation index (TSIslope) compared to G2 (1.3 ± 0.1 vs. 1.0 ± 0.2, P = 0.010), but without differences in the speed of recovery of deoxyhemoglobin [(HHb) slope], or in the magnitude of post-occlusive hyperemia (PORH). Besides TSI reoxygenation speed, G2 and G3 elicit comparable resting hemodynamic responses measured by NIRS. Thus, this study highlights the practicality and effectiveness of using relative occlusion pressures based on systolic blood pressure (SBP) rather than relying on excessively high absolute pressures.
Collapse
Affiliation(s)
- Julien Desanlis
- CIAMS, Université Paris-Saclay, Orsay, France
- CIAMS, Université d’Orléans, Orléans, France
- Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Dan Gordon
- Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Chloe French
- CIAMS, Université Paris-Saclay, Orsay, France
- CIAMS, Université d’Orléans, Orléans, France
- Cambridge Centre for Sport and Exercise Sciences, Anglia Ruskin University, Cambridge, United Kingdom
| | - Camille Calveyrac
- CIAMS, Université Paris-Saclay, Orsay, France
- CIAMS, Université d’Orléans, Orléans, France
| | - François Cottin
- CIAMS, Université Paris-Saclay, Orsay, France
- CIAMS, Université d’Orléans, Orléans, France
| | - Marie Gernigon
- CIAMS, Université Paris-Saclay, Orsay, France
- CIAMS, Université d’Orléans, Orléans, France
| |
Collapse
|
47
|
Ahmad A, Kim SJ, Jeong YJ, Khan MS, Park J, Lee DW, Lee C, Choi YJ, Yi HG. Coaxial bioprinting of a stentable and endothelialized human coronary artery-sized in vitro model. J Mater Chem B 2024; 12:8633-8646. [PMID: 39119756 DOI: 10.1039/d4tb00601a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Atherosclerosis accounts for two-thirds of deaths attributed to cardiovascular diseases, which continue to be the leading cause of mortality. Current clinical management strategies for atherosclerosis, such as angioplasty with stenting, face numerous challenges, including restenosis and late thrombosis. Smart stents, integrated with sensors that can monitor cardiovascular health in real-time, are being developed to overcome these limitations. This development necessitates rigorous preclinical trials on either animal models or in vitro models. Despite efforts being made, a suitable human-scale in vitro model compatible with a cardiovascular stent has remained elusive. To address this need, this study utilizes an in-bath bioprinting method to create a human-scale, freestanding in vitro model compatible with cardiovascular stents. Using a coaxial nozzle, a tubular structure of human coronary artery (HCA) size is bioprinted with a collagen-based bioink, ensuring good biocompatibility and suitable rheological properties for printing. We precisely replicated the dimensions of the HCA, including its internal diameter and wall thickness, and simulated the vascular barrier functionality. To simplify post-processing, a pumpless perfusion bioreactor is fabricated to culture a HCA-sized model, eliminating the need for a peristaltic pump and enabling scalability for high-throughput production. This model is expected to accelerate stent development in the future.
Collapse
Affiliation(s)
- Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Republic of Korea
| | - Seon-Jin Kim
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, 37666, Republic of Korea
| | - Yun-Jin Jeong
- Department of Automatic System, Chosun College of Science & Technology, Gwangju, 61453, Republic of Korea
| | - Muhammad Soban Khan
- Department of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jinsoo Park
- Department of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Dong-Weon Lee
- Department of Mechanical Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Changho Lee
- Department of Artificial Intelligence Convergence, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Nuclear Medicine, Chonnam National University Medical School and Hwasun Hospital, 58128, Republic of Korea
| | - Yeong-Jin Choi
- Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon, 51508, Republic of Korea.
- Advanced Materials Engineering, Korea National University of Science and Technology (UST), Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea.
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Republic of Korea
| |
Collapse
|
48
|
Bishop A, Romero JC, Tonapi S, Parihar M, Loranc E, Miller H, Lawrence L, Bassani N, Robledo D, Cao L, Nie J, Kanda K, Stoja A, Garcia N, Gorthi A, Stoveken B, Lane A, Fan T, Cassel T, Zha S, Musi N. ATM phosphorylation of CD98HC increases antiporter membrane localization and prevents chronic toxic glutamate accumulation in Ataxia telangiectasia. RESEARCH SQUARE 2024:rs.3.rs-4947457. [PMID: 39281865 PMCID: PMC11398575 DOI: 10.21203/rs.3.rs-4947457/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Ataxia telangiectasia (A-T) is a rare genetic disorder characterized by neurological defects, immunodeficiency, cancer predisposition, radiosensitivity, decreased blood vessel integrity, and diabetes. ATM, the protein mutated in A-T, responds to DNA damage and oxidative stress, but its functional relationship to the progressive clinical manifestation of A-T is not understood. CD98HC chaperones cystine/glutamate (x c - ) and cationic/neutral amino acid (y + L) antiporters to the cell membrane, and CD98HC phosphorylation by ATM accelerates membrane localization to acutely increase amino acid transport. Loss of ATM impacts tissues reliant on SLC family antiporters relevant to A-T phenotypes, such as endothelial cells (telangiectasia) and pancreatic α-cells (fatty liver and diabetes) with toxic glutamate accumulation. Bypassing the antiporters restores intracellular metabolic balance both in ATM-deficient cells and mouse models. These findings provide new insight into the long-known benefits of N-acetyl cysteine to A-T cells beyond oxidative stress through removing excess glutamate by production of glutathione.
Collapse
|
49
|
Zhang Y, Chai S, Dai H, Chen X, Meng Z, Ying X. Vascular endothelial function and its response to moderate-intensity aerobic exercise in trained and untrained healthy young men. Sci Rep 2024; 14:20450. [PMID: 39242762 PMCID: PMC11379850 DOI: 10.1038/s41598-024-71471-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024] Open
Abstract
30 min of moderate-intensity aerobic exercise per day is recommended, but the response and adaptation of endothelial function (EF) to this exercise remains controversial. The purpose of this study was to determine the changes in EF in endurance trained and untrained individuals before and after this exercise and to compare the differences between trained and untrained individuals. Twelve endurance-trained male college athletes (trained group) and 12 untrained male college students (untrained group) performed a 30-min run at an intensity of 60% VO2max. Brachial artery flow-mediated dilation (FMD) was measured before exercise, 30 min and 60 min after exercise, and the following morning. Resting diameter and maximum diameter showed large time effects (p < 0.001, η2 = 0.533; p < 0.001, η2 = 0.502). Resting diameters at 30 and 60 min after exercise were higher than before exercise in both the untrained and trained groups (p < 0.05), and maximum diameters at 30 min after exercise were higher than before exercise in both the untrained and trained groups (p < 0.01). Resting diameter and maximum diameter also exhibited some group effects (p = 0.055, η2 = 0.157; p = 0.041, η2 = 0.176). Resting diameters and maximum diameters were higher in the trained group than in the untrained group before exercise (p < 0.05). FMD (%) showed no time, group, or time-group interaction effects. 30 min of moderate-intensity aerobic exercise can increase resting and maximal arterial diameters in both trained and untrained young men, but has no effect on FMD. Long-term endurance training has the potential to increase resting and maximal arterial diameters in young men, but not necessarily FMD.
Collapse
Affiliation(s)
- Yong Zhang
- School of Medicine, Shaoxing University, Shaoxing, China
| | - Shiyi Chai
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Hailun Dai
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Xiaofei Chen
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China
| | - Zhaofeng Meng
- College of Physical Education and Health Sciences, Zhejiang Normal University, Jinhua, China.
| | - Xiaofang Ying
- School of Business, Shaoxing University, Shaoxing, China.
| |
Collapse
|
50
|
Wixler V, Zaytsev IZ, Boergeling Y, Ludwig S. The anti-inflammatory and tolerogenic potential of small spleen peptides. Front Immunol 2024; 15:1449657. [PMID: 39286259 PMCID: PMC11402738 DOI: 10.3389/fimmu.2024.1449657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
Maintaining peripheral immune tolerance and preventing harmful autoimmune reactions is a fundamental task of the immune system. However, these essential functions are significantly compromised during autoimmune disorders, creating a major challenge in treating these conditions. In this context, we provide an overview of research on small spleen polypeptides (SSPs) that naturally regulate peripheral immune tolerance. Alongside outlining the observed effects of SSPs, we summarize here the findings on the cellular and molecular mechanisms that underlie their regulatory impact. Specifically, SSPs have demonstrated remarkable effectiveness in halting the progression of developing or established autoimmune disorders like psoriasis or arthritis in animal models. They primarily target dendritic cells (DCs), swiftly prompting the production of extracellular ATP, which is then degraded and sensed by adenosine receptors. This process triggers the mTOR signaling cascade, similar to powerful immune triggers, but instead of a rapid and intense reaction, it leads to a moderate yet significant activation of the mTOR signaling cascade. This induces a tolerogenic state in dendritic cells, ultimately leading to the generation of Foxp3+ immunosuppressor Treg cells. In addition, SSPs may indirectly attenuate the autoimmune response by reducing extracellular ATP synthesis in non-immune cells, such as endothelial cells, when exposed to elevated levels of proinflammatory cytokines. SSPs thus have the potential to contribute to the restoration of peripheral immune tolerance and may offer valuable therapeutic benefits in treating autoimmune diseases.
Collapse
Affiliation(s)
- Viktor Wixler
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, Westfaelische Wilhelms-University, Muenster, Germany
| | - Igor Z Zaytsev
- Institute of Pharmaceutical Technologies, Moscow, Russia
| | - Yvonne Boergeling
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, Westfaelische Wilhelms-University, Muenster, Germany
| | - Stephan Ludwig
- Institute of Molecular Virology, Centre for Molecular Biology of Inflammation, Westfaelische Wilhelms-University, Muenster, Germany
| |
Collapse
|