1
|
He JH, Li XJ, Wang SP, Guo X, Chu HX, Xu HC, Wang YS. Eugenol Inhibits Ox-LDL-Induced Proliferation and Migration of Human Vascular Smooth Muscle Cells by Inhibiting the Ang II/MFG-E8/MCP-1 Signaling Cascade. J Inflamm Res 2024; 17:641-653. [PMID: 38328560 PMCID: PMC10847669 DOI: 10.2147/jir.s446960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 01/20/2024] [Indexed: 02/09/2024] Open
Abstract
Objective In this study, we investigated the effect and mechanism of action of eugenol on oxidized low-density lipoprotein (ox-LDL)-induced abnormal proliferation and migration of human vascular smooth muscle cells (HVSMCs). Methods HVSMCs were treated with 100 ug/mL ox-LDL for 24 hours to establish a cell model. After 1-hour pretreatment, eugenol at concentrations of 5, 25, and 50 uM was added. Cell viability was assessed using an MTT assay, PCNA expression was detected using Western blot, cell cycle distribution was analyzed using flow cytometry, and cell migration ability was evaluated using wound healing and Transwell migration assays. To investigate the mechanisms, Ang II receptors were inhibited by 1000 nM valsartan, MFG-E8 was knocked down by shRNA, MCP-1 was inhibited by siRNA, and MFG-E8 was overexpressed using plasmids. Results The findings from this study elucidated the stimulatory impact of ox-LDL on the proliferation and functionality of HVSMCs. Different concentrations of eugenol effectively mitigated the enhanced activity of HVSMCs induced by ox-LDL, with 50 uM eugenol exhibiting the most pronounced inhibitory effect. Flow cytometry and Western blot results showed ox-LDL reduced G1 phase cells and increased PCNA expression, while 50 uM eugenol inhibited ox-LDL-induced HVSMC proliferation. In wound healing and Transwell migration experiments, the ox-LDL group showed larger cell scratch filling and migration than the control group, both of which were inhibited by 50 uM eugenol. Inhibiting the Ang II/MFG-E8/MCP-1 signaling cascade mimicked eugenol's effects, while MFG-E8 overexpression reversed eugenol's inhibitory effect. Conclusion Eugenol can inhibit the proliferation and migration of ox-LDL-induced HVSMCs by inhibiting Ang II/MFG-E8/MCP-1 signaling cascade, making it a potential therapeutic drug for atherosclerosis.
Collapse
Affiliation(s)
- Jia-Huan He
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 13000, People’s Republic of China
| | - Xiang-Jun Li
- Department of Experimental Pharmacology and Toxicology, College of Pharmacy, Jilin University, Changchun, 130000, People’s Republic of China
| | - Shi-Peng Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 13000, People’s Republic of China
| | - Xia Guo
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 13000, People’s Republic of China
| | - Hao-Xuan Chu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 13000, People’s Republic of China
| | - Han-Chi Xu
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 13000, People’s Republic of China
| | - Yu-Shi Wang
- Department of Cardiology, The First Hospital of Jilin University, Changchun, 13000, People’s Republic of China
| |
Collapse
|
2
|
Massamba VK, Talbot D, Milot A, Trudel X, Dionne CE, Vézina M, Mâsse B, Gilbert-Ouimet M, Dagenais GR, Pearce N, Brisson C. Association between psychosocial work-related factors at midlife and arterial stiffness at older age in a prospective cohort of 1736 white-collar workers. BMJ Open 2023; 13:e073649. [PMID: 37758677 PMCID: PMC10537828 DOI: 10.1136/bmjopen-2023-073649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
OBJECTIVE Arterial stiffness and exposure to psychosocial work-related factors increase the risk of developing cardiovascular disease. However, little is known about the relationship between psychosocial work-related factors and arterial stiffness. We aimed to examine this relationship. DESIGN Prospective cohort study. SETTING Public organisations in Quebec City, Canada. PARTICIPANTS The study included 1736 white-collar workers (women 52%) from 19 public organisations. PRIMARY AND SECONDARY OUTCOME MEASURES Association between psychosocial work-related factors from the job strain and effort-reward imbalance (ERI) models assessed at study baseline (1999-2001) with validated instruments and arterial stiffness assessed using carotid-femoral pulse wave velocity at follow-up, on average 16 years later (2015-2018). Generalised estimating equations were used to estimate differences in arterial stiffness between exposed and unexposed participants. Subgroup analyses according to sex, age, blood pressure (BP), cardiovascular risk score and employment status were conducted. RESULTS Among participants with high diastolic BP (≥90 mm Hg) at baseline, aged 47 on average, those exposed to high job strain had higher arterial stiffness (1.38 m/s (95% CI: 0.57 to 2.19)) at follow-up, 16 years later, following adjustment for a large set of potential confounders. The trend was similar in participants with high systolic BP (≥140 mm Hg) exposed to high job strain (0.84 m/s (95% CI: -0.35 to 2.03)). No association was observed for ERI in the total sample and counterintuitive associations were observed in subgroup analyses. CONCLUSIONS Job strain may have a long-term deleterious effect on arterial stiffness in people with high BP. Interventions at midlife to reduce job strain may mitigate arterial stiffness progression.
Collapse
Affiliation(s)
- Victoria K Massamba
- Department of Social and Preventive Medicine, Université Laval, Quebec, Quebec, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
| | - Denis Talbot
- Department of Social and Preventive Medicine, Université Laval, Quebec, Quebec, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
| | - Alain Milot
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
- Department of Medicine, Université Laval, Quebec, Quebec, Canada
| | - Xavier Trudel
- Department of Social and Preventive Medicine, Université Laval, Quebec, Quebec, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
| | - Clermont E Dionne
- Department of Social and Preventive Medicine, Université Laval, Quebec, Quebec, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
| | - Michel Vézina
- Institut national de sante publique du Quebec, Quebec, Quebec, Canada
| | - Benoit Mâsse
- Department of Social and Preventive Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Mahée Gilbert-Ouimet
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
- Department of Health Sciences, Université du Québec à Rimouski, Rimouski, Quebec, Canada
| | - Gilles R Dagenais
- Institut universitaire de cardiologie et de pneumologie de Quebec, Quebec, Quebec, Canada
| | - Neil Pearce
- Departments of Medical Statistics and Non-communicable Disease Epidemiology, London School of Hygiene and Tropical Medicine, London, UK
| | - Chantal Brisson
- Department of Social and Preventive Medicine, Université Laval, Quebec, Quebec, Canada
- Centre de recherche du CHU de Québec-Université Laval, Quebec, Quebec, Canada
| |
Collapse
|
3
|
Menon R, Otto EA, Barisoni L, Melo Ferreira R, Limonte CP, Godfrey B, Eichinger F, Nair V, Naik AS, Subramanian L, D'Agati V, Henderson JM, Herlitz L, Kiryluk K, Moledina DG, Moeckel GW, Palevsky PM, Parikh CR, Randhawa P, Rosas SE, Rosenberg AZ, Stillman I, Toto R, Torrealba J, Vazquez MA, Waikar SS, Alpers CE, Nelson RG, Eadon MT, Kretzler M, Hodgin JB. Defining the molecular correlate of arteriolar hyalinosis in kidney disease progression by integration of single cell transcriptomic analysis and pathology scoring. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.14.23291150. [PMID: 37398386 PMCID: PMC10312894 DOI: 10.1101/2023.06.14.23291150] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Arteriolar hyalinosis in kidneys is an independent predictor of cardiovascular disease, the main cause of mortality in chronic kidney disease (CKD). The underlying molecular mechanisms of protein accumulation in the subendothelial space are not well understood. Using single cell transcriptomic data and whole slide images from kidney biopsies of patients with CKD and acute kidney injury in the Kidney Precision Medicine Project, the molecular signals associated with arteriolar hyalinosis were evaluated. Co-expression network analysis of the endothelial genes yielded three gene set modules as significantly associated with arteriolar hyalinosis. Pathway analysis of these modules showed enrichment of transforming growth factor beta / bone morphogenetic protein (TGFβ / BMP) and vascular endothelial growth factor (VEGF) signaling pathways in the endothelial cell signatures. Ligand-receptor analysis identified multiple integrins and cell adhesion receptors as over-expressed in arteriolar hyalinosis, suggesting a potential role of integrin-mediated TGFβ signaling. Further analysis of arteriolar hyalinosis associated endothelial module genes identified focal segmental glomerular sclerosis as an enriched term. On validation in gene expression profiles from the Nephrotic Syndrome Study Network cohort, one of the three modules was significantly associated with the composite endpoint (> 40% reduction in estimated glomerular filtration rate (eGFR) or kidney failure) independent of age, sex, race, and baseline eGFR, suggesting poor prognosis with elevated expression of genes in this module. Thus, integration of structural and single cell molecular features yielded biologically relevant gene sets, signaling pathways and ligand-receptor interactions, underlying arteriolar hyalinosis and putative targets for therapeutic intervention.
Collapse
|
4
|
Mansur AJ. SAGE Score and Pulse Wave Velocity in Non-Hypertensive Individuals. Arq Bras Cardiol 2023; 120:e20220881. [PMID: 36856246 PMCID: PMC10263455 DOI: 10.36660/abc.20220881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Affiliation(s)
- Alfredo José Mansur
- Instituto do CoraçãoFaculdade de MedicinaUniversidade de São PauloSão PauloSPBrasilInstituto do Coração da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP – Brasil
| |
Collapse
|
5
|
Chintapula U, Chikate T, Sahoo D, Kieu A, Guerrero Rodriguez ID, Nguyen KT, Trott D. Immunomodulation in age-related disorders and nanotechnology interventions. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2023; 15:e1840. [PMID: 35950266 PMCID: PMC9840662 DOI: 10.1002/wnan.1840] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 05/19/2022] [Accepted: 06/01/2022] [Indexed: 01/31/2023]
Abstract
Recently, the aging population has increased exponentially around the globe bringing more challenges to improve quality of life in those populations while reducing the economic burden on healthcare systems. Aging is associated with changes in the immune system culminating in detrimental effects such as immune dysfunction, immunosenescence, and chronic inflammation. Age-related decline of immune functions is associated with various pathologies including cardiovascular, autoimmune, neurodegenerative, and infectious diseases to name a few. Conventional treatment addresses the onset of age-related diseases by early detection of risk factors, administration of vaccines as preventive care, immunomodulatory treatment, and other dietary supplements. However, these approaches often come with systemic side-effects, low bioavailability of therapeutic agents, and poor outcomes seen in the elderly. Recent innovations in nanotechnology have led to the development of novel biomaterials/nanomaterials, which explore targeted drug delivery and immunomodulatory interactions in vivo. Current nanotechnology-based immunomodulatory approaches that have the potential to be used as therapeutic interventions for some prominent age-related diseases are discussed here. Finally, we explore challenges and future aspects of nanotechnology in the treatments of age-related disorders to improve quality of life in the elderly. This article is categorized under: Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Uday Chintapula
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Tanmayee Chikate
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Deepsundar Sahoo
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | - Amie Kieu
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
| | | | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas, USA
- Joint Bioengineering Program, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel Trott
- Department of Kinesiology, University of Texas at Arlington, Arlington, Texas, USA
| |
Collapse
|
6
|
Romagnolo D, D′Avila A, Migliore F, Vergara P. Editorial: Sport activity: From beneficial effects to cardiac disease. Front Physiol 2022; 13:1094048. [PMID: 36483294 PMCID: PMC9724559 DOI: 10.3389/fphys.2022.1094048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 11/16/2022] [Indexed: 06/23/2024] Open
Affiliation(s)
- Davide Romagnolo
- IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Andre D′Avila
- Beth Israel Deaconess Medical Center, Harvard Medical School, Harvard-Thorndike Electrophysiology Institute, Boston, MA, United States
| | - Federico Migliore
- Department of Cardiac, Thoracic and Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Pasquale Vergara
- Cardiac Electrophysiology and Arrhythmology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
7
|
Oliveira AC, Cunha PMGM, Vitorino PVDO, Souza ALL, Deus GD, Feitosa A, Barbosa ECD, Gomes MM, Jardim PCBV, Barroso WKS. Vascular Aging and Arterial Stiffness. Arq Bras Cardiol 2022; 119:604-615. [PMID: 36287415 PMCID: PMC9563886 DOI: 10.36660/abc.20210708] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 05/11/2022] [Indexed: 11/18/2022] Open
Abstract
O envelhecimento biológico é reflexo da interação entre genética, idade cronológica e fatores externos; é a base para novos conceitos em envelhecimento vascular, cuja progressão é determinada pela diferença entre idade biológica e cronológica. Do ponto de vista estrutural, os efeitos do envelhecimento vascular são mais evidentes na camada média das grandes artérias elásticas e resultam em aumento da rigidez arterial, da dilatação do lúmen e da espessura da parede. Esses efeitos são descritos no continuum de envelhecimento cardiovascular (proposto por Dzau em 2010) em que as etapas progressivas de lesões da microvasculatura de coração, rins e cérebro, têm início a partir do processo de envelhecimento. O aumento da rigidez arterial pode ser verificado de forma não invasiva por vários métodos. Os eventos cardiovasculares têm sido tradicionalmente previstos utilizando escores que combinam fatores de risco convencionais para aterosclerose. No continuum cardiovascular clássico (Dzau, 2006), é desafiador avaliar o peso exato da contribuição de cada fator de risco; entretanto, por refletir o dano precoce e cumulativo desses fatores de riscos cardiovascular, a rigidez arterial reflete o verdadeiro dano à parede arterial. Este artigo fornece uma visão geral dos mecanismos da fisiopatogenia, alterações estruturais das artérias e consequências hemodinâmicas do envelhecimento arterial; métodos não invasivos para a avaliação da rigidez arterial e da medida central da pressão arterial; o continuum de envelhecimento cardiovascular, e aplicação do conceito de rigidez arterial na estratificação de risco cardiovascular.
Collapse
Affiliation(s)
| | | | | | - Ana Luiza Lima Souza
- Universidade Federal de Goiás , Liga de Hipertensão , Goiânia , GO - Brasil.,Universidade Federal de Goiás - Programa de Pós-Graduação em Ciências da Saúde , Goiânia , GO - Brasil
| | - Gilcimar Divino Deus
- Pontifícia Universidade Católica de Goiás - Escola de Ciências Exatas e da Computação , Goiânia , GO - Brasil
| | - Audes Feitosa
- Universidade de Pernambuco , Recife , PE - Brasil.,Universidade Católica de Pernambuco , Recife , PE - Brasil
| | | | - Marco Mota Gomes
- Centro Universitario CESMAC - Hospital do Coração , Maceió , AL - Brasil
| | | | | |
Collapse
|
8
|
Peták F, Kovács BN, Agócs S, Virág K, Nyári T, Molnár A, Südy R, Lengyel C, Babik B. Seasonal changes in proportion of cardiac surgeries associated with diabetes, smoking and elderly age. PLoS One 2022; 17:e0274105. [PMID: 36136994 PMCID: PMC9498963 DOI: 10.1371/journal.pone.0274105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 08/22/2022] [Indexed: 11/25/2022] Open
Abstract
Background Seasonal variations in the ambient temperature may affect the exacerbation of cardiovascular diseases. Our primary objective was to evaluate the seasonality of the monthly proportion of cardiac surgeries associated with diabetes, smoking and/or elderly age at a tertiary-care university hospital in East-Central Europe with a temperate climate zone. As a secondary objective, we also assessed whether additional factors affecting small blood vessels (smoking, aging, obesity) modulate the seasonal variability of diabetes. Methods Medical records were analyzed for 9838 consecutive adult patients who underwent cardiac surgery in 2007–2018. Individual seasonal variations of diabetes, smoking, and elderly patients were analyzed monthly, along with the potential risk factors for cardiovascular complication. We also characterized whether pairwise coexistence of diabetes, smoking, and elderly age augments or blunts the seasonal variations. Results Seasonal variations in the monthly proportion of cardiac surgeries associated with diabetes, smoking and/or elderly age were observed. The proportion of cardiac surgeries of non-elderly and smoking patients with diabetes peaked in winter (amplitude of change as [peak-nadir]/nadir: 19.2%, p<0.02), which was associated with increases in systolic (6.1%, p<0.001) and diastolic blood pressures (4.4%, p<0.05) and serum triglyceride levels (27.1%, p<0.005). However, heart surgery in elderly patients without diabetes and smoking was most frequently required in summer (52.1%, p<0.001). Concomitant occurrence of diabetes and smoking had an additive effect on the requirement for cardiac surgery (107%, p<0.001), while the simultaneous presence of older age and diabetes or smoking eliminated seasonal variations. Conclusions Scheduling regular cardiovascular control in accordance with periodicities in diabetes, elderly, and smoking patients more than once a year may improve patient health and social consequences. Trial registration NCT03967639.
Collapse
Affiliation(s)
- Ferenc Peták
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- * E-mail:
| | - Barbara N. Kovács
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- Department of Anesthesiology and Intensive Therapy, University of Szeged, Szeged, Hungary
| | - Szilvia Agócs
- Department of Anesthesiology and Intensive Therapy, University of Szeged, Szeged, Hungary
- Department of Internal Medicine and Cardiology Center, Cardiac Surgery Unit, University of Szeged, Szeged, Hungary
| | - Katalin Virág
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Tibor Nyári
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
| | - Andrea Molnár
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- Department of Anesthesiology and Intensive Therapy, University of Szeged, Szeged, Hungary
| | - Roberta Südy
- Department of Medical Physics and Informatics, University of Szeged, Szeged, Hungary
- Department of Anesthesiology and Intensive Therapy, University of Szeged, Szeged, Hungary
| | - Csaba Lengyel
- Department of Internal Medicine, University of Szeged, Szeged, Hungary
| | - Barna Babik
- Department of Anesthesiology and Intensive Therapy, University of Szeged, Szeged, Hungary
- Department of Internal Medicine and Cardiology Center, Cardiac Surgery Unit, University of Szeged, Szeged, Hungary
| |
Collapse
|
9
|
Fabiani M, Asnakew BA, Bowie DC, Chism SM, Clements GM, Gardner JC, Islam SS, Rubenstein SL, Gratton G. A healthy mind in a healthy body: Effects of arteriosclerosis and other risk factors on cognitive aging and dementia. THE PSYCHOLOGY OF LEARNING AND MOTIVATION 2022; 77:69-123. [PMID: 37139101 PMCID: PMC10153623 DOI: 10.1016/bs.plm.2022.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
In this review we start from the assumption that, to fully understand cognitive aging, it is important to embrace a holistic view, integrating changes in bodily, brain, and cognitive functions. This broad view can help explain individual differences in aging trajectories and could ultimately enable prevention and remediation strategies. As the title of this review suggests, we claim that there are not only indirect but also direct effects of various organ systems on the brain, creating cascades of phenomena that strongly contribute to age-related cognitive decline. Here we focus primarily on the cerebrovascular system, because of its direct effects on brain health and close connections with the development and progression of Alzheimer's Disease and other types of dementia. We start by reviewing the main cognitive changes that are often observed in normally aging older adults, as well as the brain systems that support them. Second, we provide a brief overview of the cerebrovascular system and its known effects on brain anatomy and function, with a focus on aging. Third, we review genetic and lifestyle risk factors that may affect the cerebrovascular system and ultimately contribute to cognitive decline. Lastly, we discuss this evidence, review limitations, and point out avenues for additional research and clinical intervention.
Collapse
Affiliation(s)
- Monica Fabiani
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Bethlehem A. Asnakew
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Daniel C. Bowie
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Sydney M. Chism
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Grace M. Clements
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Jennie C. Gardner
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Samia S. Islam
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Samantha L. Rubenstein
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| | - Gabriele Gratton
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, IL, United States
- Psychology Department, University of Illinois at Urbana-Champaign, Champaign, IL, United States
| |
Collapse
|
10
|
Vascular calcification in different arterial beds in ex vivo ring culture and in vivo rat model. Sci Rep 2022; 12:11861. [PMID: 35831341 PMCID: PMC9279329 DOI: 10.1038/s41598-022-15739-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 06/28/2022] [Indexed: 11/08/2022] Open
Abstract
Vascular calcification is a risk factor for cardiovascular and kidney diseases. Medial calcification may differently affect the arterial tree depending on vessel location and smooth muscle injury. The aim was to map the anatomical distribution of vascular calcifications on different arteries and artery locations, in cultured artery rings (ex vivo) and in a rat model of elastocalcinosis (in vivo). Vascular calcification was assessed histologically (von Kossa staining of the media) and by calcium content measurement. Arteries of different sizes were harvested from untreated rats for ring culture and from the vitamin D3-nicotine (VDN) rat model for direct observation. When cultured in pro-calcifying conditions, thoracic aorta exhibited similar calcification from the arch to the diaphragm. Calcification increased in abdominal aorta along with the reduction in cross sectional area. Carotid and renal arteries exhibited similar ex vivo calcification. In VDN rats, calcification was greater in carotid artery than in aorta, and was accompanied by fibrosis and apoptosis. Ex vivo, calcification was increased by the induction of lesions on arteries. Along the vascular tree, calcification of the arterial wall increases with the narrowing of vessels in ex vivo ring culture and in vivo. The observed differences represent local susceptibility of the vessels to the calcifying processes.
Collapse
|
11
|
Laučytė-Cibulskienė A, Smaliukaitė M, Dadonienė J, Čypienė A, Mikolaitytė J, Ryliškytė L, Laucevičius A, Badarienė J. Inflammaging and Vascular Function in Metabolic Syndrome: The Role of Hyperuricemia. Medicina (B Aires) 2022; 58:medicina58030373. [PMID: 35334550 PMCID: PMC8953262 DOI: 10.3390/medicina58030373] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 02/24/2022] [Accepted: 02/28/2022] [Indexed: 01/10/2023] Open
Abstract
Background and Objectives: Early vascular aging determines a more rapid course of age-related arterial changes. It may be induced by a proinflammatory state, caused by hyperuricemia and metabolic syndrome and their interrelationship. However, the impact of serum uric acid (SUA) on early arterial stiffening and vascular function remains uncertain. Materials and Methods: A total of 696 participants (439 women aged 50–65 and 257 men aged 40–55) from the Lithuanian High Cardiovascular Risk (LitHiR) primary prevention program were enrolled in the study. They underwent anthropometric measurements and laboratory testing along with arterial parameters’ evaluation. Quality carotid stiffness (QCS), carotid-radial pulse wave velocity (crPWV), carotid-femoral pulse wave velocity (cfPWV), flow-mediated dilatation (FMD), and carotid intima-media thickness (CIMT) were registered. Results: We found that hyperuricemia was significantly associated with inflammation, registered by high-sensitivity C-reactive protein in both sexes. A very weak but significant association was observed between cfPWV and SUA in men and in women, while, after adjusting for risk factors, it remained significant only in women. A positive, weak, but significant association was also observed for QCS, both right and left in women. No relationship was observed between crPWV, FMD, CIMT, and SUA.
Collapse
Affiliation(s)
- Agnė Laučytė-Cibulskienė
- Department of Clinical Sciences Malmo, Lund University, Skane University Hospital, 205 02 Malmö, Sweden;
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
| | - Monika Smaliukaitė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
- Correspondence:
| | - Jolanta Dadonienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania;
| | - Alma Čypienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania;
| | - Jurgita Mikolaitytė
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania;
| | - Ligita Ryliškytė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
| | - Aleksandras Laucevičius
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania;
| | - Jolita Badarienė
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania; (J.D.); (A.Č.); (L.R.); (A.L.); (J.B.)
- State Research Institute Centre for Innovative Medicine, 08406 Vilnius, Lithuania;
| |
Collapse
|
12
|
Changed pathophysiology of thoracic aorta after aortic arch repair. J Vasc Surg 2022; 75:1122-1123. [DOI: 10.1016/j.jvs.2021.11.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/05/2021] [Indexed: 11/19/2022]
|
13
|
El Fol A, Ammar W, Sharaf Y, Youssef G. The central arterial stiffness parameters in decompensated versus compensated states of heart failure: a paired comparative cohort study. Egypt Heart J 2022; 74:2. [PMID: 34978636 PMCID: PMC8724513 DOI: 10.1186/s43044-021-00236-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 12/22/2021] [Indexed: 11/10/2022] Open
Abstract
Background Arterial stiffness is strongly linked to the pathogenesis of heart failure and the development of acute decompensation in patients with stable chronic heart failure. This study aimed to compare arterial stiffness indices in patients with heart failure with reduced ejection fraction (HFrEF) during the acute decompensated state, and three months later after hospital discharge during the compensated state. Results One hundred patients with acute decompensated HFrEF (NYHA class III and IV) and left ventricular ejection fraction ≤ 35% were included in the study. During the initial and follow-up visits, all patients underwent full medical history taking, clinical examination, transthoracic echocardiography, and non-invasive pulse wave analysis by the Mobil-O-Graph 24-h device for measurement of arterial stiffness. The mean age was 51.6 ± 6.1 years and 80% of the participants were males. There was a significant reduction of the central arterial stiffness indices in patients with HFrEF during the compensated state compared to the decompensated state. During the decompensated state, patients presented with NYHA FC IV (n = 64) showed higher AI (24.5 ± 10.0 vs. 16.8 ± 8.6, p < 0.001) and pulse wave velocity (9.2 ± 1.3 vs. 8.5 ± 1.2, p = 0.021) than patients with NYHA FC III, and despite the relatively smaller number of females, they showed higher stiffness indices than males. Conclusions Central arterial stiffness indices in patients with HFrEF were significantly lower in the compensated state than in the decompensated state. Patients with NYHA FC IV and female patients showed higher stiffness indices in their decompensated state of heart failure.
Collapse
Affiliation(s)
- Ahmed El Fol
- Cardiovascular Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Waleed Ammar
- Cardiovascular Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Yasser Sharaf
- Cardiovascular Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ghada Youssef
- Cardiovascular Department, Faculty of Medicine, Cairo University, Cairo, Egypt.
| |
Collapse
|
14
|
Kozakova M, Palombo C. Vascular Ageing and Aerobic Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:10666. [PMID: 34682413 PMCID: PMC8535583 DOI: 10.3390/ijerph182010666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 12/15/2022]
Abstract
Impairment of vascular function, in particular endothelial dysfunction and large elastic artery stiffening, represents a major link between ageing and cardiovascular risk. Clinical and experimental studies identified numerous mechanisms responsible for age-related decline of endothelial function and arterial compliance. Since most of these mechanisms are related to oxidative stress or low-grade inflammation, strategies that suppress oxidative stress and inflammation could be effective for preventing age-related changes in arterial function. Indeed, aerobic physical activity, which has been shown to improve intracellular redox balance and mitochondrial health and reduce levels of systemic inflammatory markers, also improves endothelial function and arterial distensibility and reduces risk of cardiovascular diseases. The present paper provides a brief overview of processes underlying age-related changes in arterial function, as well as the mechanisms through which aerobic exercise might prevent or interrupt these processes, and thus attenuate vascular ageing.
Collapse
Affiliation(s)
- Michaela Kozakova
- Department of Clinical and Experimental Medicine, University of Pisa, 56124 Pisa, Italy;
| | - Carlo Palombo
- Department of Surgical, Medical, Molecular Pathology and Critical Care Medicine, University of Pisa, 56124 Pisa, Italy
| |
Collapse
|
15
|
Martínez-Ceballos MA, Sinning Rey JC, Alzate-Granados JP, Mendoza-Pinto C, García-Carrasco M, Montes-Zabala L, Vargas-Vergara D, Munguia-Realpozo P, Etchegaray-Morales I, Rojas-Villarraga A. Coronary calcium in autoimmune diseases: A systematic literature review and meta-analysis. Atherosclerosis 2021; 335:68-76. [PMID: 34592584 DOI: 10.1016/j.atherosclerosis.2021.09.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 08/29/2021] [Accepted: 09/16/2021] [Indexed: 01/16/2023]
Abstract
BACKGROUND AND AIMS Autoimmune diseases (AID) share various clinical signs and symptoms and pathophysiological mechanisms including the increased risk of cardiovascular disease. The coronary artery calcium score (CACS) is potentially useful in improving the cardiovascular risk assessment. The aim of this study was to evaluate CACS in six AIDs analyzed as a group compared with controls through a systematic literature review (SLR) and meta-analysis. METHODS A literature search (Medline/OVID, Lilacs, Embase, and Cochrane/OVID) up to January 6, 2021 was made (PROSPERO CRD42020197182). Observational studies (patients with six AIDs: rheumatoid arthritis [RA], systemic lupus erythematosus [SLE], Sjögren's syndrome, systemic sclerosis, dermatopolymyositis, and antiphospholipid syndrome) compared with controls were included. CACS, reported in Agatston units, was the primary outcome in both groups. Mean differences and a random-effects model (DerSimonian and Laird) were calculated. RESULTS Nineteen articles were meta-analyzed (4568 subjects: 2142 AID and 2426 controls). Mean age was 48.1 and 44.2 years, respectively and 75.6% and 84.9% were women, respectively. Of cases, 52.9% had RA, 44.4% SLE and 2.7% had systemic sclerosis. The pooled analysis showed a higher CACS in patients with AIDs (7.42; 95% CI 1.79 to 13.05; chi2-p = 0.01) compared with controls. Meta-regression models showed that age in cases and controls reduced the difference in CACS between groups (p < 0.05), HDL had an inverse relationship (p = 0.04), and CRP levels had a directly proportional relationship with CACS in cases (p = 0.036). CONCLUSIONS The quantitative results of this meta-analysis suggest that CACS is higher in patients with AID, possibly due to chronic exposure to pro-inflammatory molecules. These results have clinical implications since the finding of highly elevated CACS in patients with AID will enable physicians and researchers to develop a risk stratification model that includes CACS as one of the screening tools for detecting coronary atherosclerosis in these patients.
Collapse
Affiliation(s)
| | - Jhoan Camilo Sinning Rey
- Department of Cardiology, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | | | - Claudia Mendoza-Pinto
- Systemic Autoimmune Diseases Research Unit, High Specialized Medical Unit, UMAE CMNMAC - CIBIOR, Mexican Social Security Institute, Puebla, Mexico; Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Mario García-Carrasco
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Lorena Montes-Zabala
- Department of Cardiology, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Diana Vargas-Vergara
- Department of Cardiology, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Colombia
| | - Pamela Munguia-Realpozo
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | - Ivet Etchegaray-Morales
- Department of Rheumatology, Medicine School, Benemérita Universidad Autónoma de Puebla, Mexico
| | | |
Collapse
|
16
|
Santos-Moreno P, Burgos-Angulo G, Martinez-Ceballos MA, Pizano A, Echeverri D, Bautista-Niño PK, Roks AJM, Rojas-Villarraga A. Inflammaging as a link between autoimmunity and cardiovascular disease: the case of rheumatoid arthritis. RMD Open 2021; 7:rmdopen-2020-001470. [PMID: 33468563 PMCID: PMC7817822 DOI: 10.1136/rmdopen-2020-001470] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2020] [Revised: 11/27/2020] [Accepted: 12/21/2020] [Indexed: 12/13/2022] Open
Abstract
Currently, traditional and non-traditional risk factors for cardiovascular disease have been established. The first group includes age, which constitutes one of the most important factors in the development of chronic diseases. The second group includes inflammation, the pathophysiology of which contributes to an accelerated process of vascular remodelling and atherogenesis in autoimmune diseases. Indeed, the term inflammaging has been used to refer to the inflammatory origin of ageing, explicitly due to the chronic inflammatory process associated with age (in healthy individuals). Taking this into account, it can be inferred that people with autoimmune diseases are likely to have an early acceleration of vascular ageing (vascular stiffness) as evidenced in the alteration of non-invasive cardiovascular tests such as pulse wave velocity. Thus, an association is created between autoimmunity and high morbidity and mortality rates caused by cardiovascular disease in this population group. The beneficial impact of the treatments for rheumatoid arthritis at the cardiovascular level has been reported, opening new opportunities for pharmacotherapy.
Collapse
Affiliation(s)
| | - Gabriel Burgos-Angulo
- Internal Medicine Department, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogota, Cundinamarca, Colombia
| | | | - Alejandro Pizano
- Vascular Function Research Laboratory and Department of Interventional Cardiology, Fundación Cardioinfantil Instituto de Cardiología, Bogota, Cundinamarca, Colombia
| | - Dario Echeverri
- Vascular Function Research Laboratory and Department of Interventional Cardiology, Fundación Cardioinfantil Instituto de Cardiología, Bogota, Cundinamarca, Colombia
| | - Paula K Bautista-Niño
- Research Center, Fundación Cardiovascular de Colombia, Floridablanca, Santander, Colombia
| | - Anton J M Roks
- Department of Internal Medicine, Division of Pharmacology and Vascular Medicine, Erasmus Medical Center Department of Internal Medicine, Rotterdam, South Holland, The Netherlands
| | - Adriana Rojas-Villarraga
- Research Division, Fundación Universitaria de Ciencias de la Salud (FUCS), Bogotá, Cundinamarca, Colombia
| |
Collapse
|
17
|
Mohammadi H, Vincent T, Peng K, Nigam A, Gayda M, Fraser S, Joanette Y, Lesage F, Bherer L. Coronary artery disease and its impact on the pulsatile brain: A functional NIRS study. Hum Brain Mapp 2021; 42:3760-3776. [PMID: 33991155 PMCID: PMC8288102 DOI: 10.1002/hbm.25463] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2021] [Revised: 04/12/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Recent studies have reported that optical indices of cerebral pulsatility are associated with cerebrovascular health in older adults. Such indices, including cerebral pulse amplitude and the pulse relaxation function (PRF), have been previously applied to quantify global and regional cerebral pulsatility. The aim of the present study was to determine whether these indices are modulated by cardiovascular status and whether they differ between individuals with low or high cardiovascular risk factors (LCVRF and HCVRF) and coronary artery disease (CAD). A total of 60 older adults aged 57-79 were enrolled in the study. Participants were grouped as LCVRF, HCVRF, and CAD. Participants were asked to walk freely on a gym track while a near-infrared spectroscopy (NIRS) device recorded hemodynamics data. Low-intensity, short-duration walking was used to test whether a brief cardiovascular challenge could increase the difference of pulsatility indices with respect to cardiovascular status. Results indicated that CAD individuals have higher global cerebral pulse amplitude compared with the other groups. Walking reduced global cerebral pulse amplitude and PRF in all groups but did not increase the difference across the groups. Instead, walking extended the spatial distribution of cerebral pulse amplitude to the anterior prefrontal cortex when CAD was compared to the CVRF groups. Further research is needed to determine whether cerebral pulse amplitude extracted from data acquired with NIRS, which is a noninvasive, inexpensive method, can provide an index to characterize the cerebrovascular status associated with CAD.
Collapse
Affiliation(s)
- Hanieh Mohammadi
- Laboratory of Optical and Molecular ImagingBiomedical Engineering Institute, Polytechnique MontrealQuebecCanada
- Research CenterUniversity Institute of Geriatrics of MontrealMontrealQuebecCanada
- Research CenterEPIC Centre of Montreal Heart InstituteMontrealQuebecCanada
| | - Thomas Vincent
- Research CenterEPIC Centre of Montreal Heart InstituteMontrealQuebecCanada
| | - Ke Peng
- Center for Pain and the BrainBoston Children's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Research CenterUniversity of Montreal Health CentreMontrealQuebecCanada
| | - Anil Nigam
- Research CenterEPIC Centre of Montreal Heart InstituteMontrealQuebecCanada
| | - Mathieu Gayda
- Research CenterEPIC Centre of Montreal Heart InstituteMontrealQuebecCanada
| | - Sarah Fraser
- Interdisciplinary School of Health Sciences, Faculty of Health SciencesUniversity of OttawaOttawaOntarioCanada
| | - Yves Joanette
- Research CenterUniversity Institute of Geriatrics of MontrealMontrealQuebecCanada
- Faculty of MedicineUniversity of MontrealMontrealQuebecCanada
| | - Frédéric Lesage
- Laboratory of Optical and Molecular ImagingBiomedical Engineering Institute, Polytechnique MontrealQuebecCanada
- Research CenterEPIC Centre of Montreal Heart InstituteMontrealQuebecCanada
| | - Louis Bherer
- Research CenterUniversity Institute of Geriatrics of MontrealMontrealQuebecCanada
- Research CenterEPIC Centre of Montreal Heart InstituteMontrealQuebecCanada
- Faculty of MedicineUniversity of MontrealMontrealQuebecCanada
| |
Collapse
|
18
|
Para I, Albu A, Porojan MD. Adipokines and Arterial Stiffness in Obesity. ACTA ACUST UNITED AC 2021; 57:medicina57070653. [PMID: 34202323 PMCID: PMC8305474 DOI: 10.3390/medicina57070653] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 06/17/2021] [Accepted: 06/21/2021] [Indexed: 12/11/2022]
Abstract
Adipokines are active molecules with pleiotropic effects produced by adipose tissue and involved in obesity-related metabolic and cardiovascular diseases. Arterial stiffness, which is a consequence of arteriosclerosis, has been shown to be an independent predictor of cardiovascular morbidity and mortality. The pathogenesis of arterial stiffness is complex but incompletely understood. Adipokines dysregulation may induce, by various mechanisms, vascular inflammation, endothelial dysfunction, and vascular remodeling, leading to increased arterial stiffness. This article summarizes literature data regarding adipokine-related pathogenetic mechanisms involved in the development of arterial stiffness, particularly in obesity, as well as the results of clinical and epidemiological studies which investigated the relationship between adipokines and arterial stiffness.
Collapse
Affiliation(s)
- Ioana Para
- 4th Department of Internal Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| | - Adriana Albu
- 2nd Department of Internal Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
- Correspondence:
| | - Mihai D. Porojan
- 2nd Department of Internal Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400012 Cluj-Napoca, Romania;
| |
Collapse
|
19
|
Kim SH, Monticone RE, McGraw KR, Wang M. Age-associated proinflammatory elastic fiber remodeling in large arteries. Mech Ageing Dev 2021; 196:111490. [PMID: 33839189 PMCID: PMC8154723 DOI: 10.1016/j.mad.2021.111490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Elastic fibers are the main components of the extracellular matrix of the large arterial wall. Elastic fiber remodeling is an intricate process of synthesis and degradation of the core elastin protein and microfibrils accompanied by the assembly and disassembly of accessory proteins. Age-related morphological, structural, and functional proinflammatory remodeling within the elastic fiber has a profound effect upon the integrity, elasticity, calcification, amyloidosis, and stiffness of the large arterial wall. An age-associated increase in arterial stiffness is a major risk factor for the pathogenesis of diseases of the large arteries such as hypertensive and atherosclerotic vasculopathy. This mini review is an update on the key molecular, cellular, functional, and structural mechanisms of elastic fiber proinflammatory remodeling in large arteries with aging. Targeting structural and functional integrity of the elastic fiber may be an effective approach to impede proinflammatory arterial remodeling with advancing age.
Collapse
Affiliation(s)
- Soo Hyuk Kim
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Robert E Monticone
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Kimberly R McGraw
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
20
|
Pauls SD, Du Y, Clair L, Winter T, Aukema HM, Taylor CG, Zahradka P. Impact of Age, Menopause, and Obesity on Oxylipins Linked to Vascular Health. Arterioscler Thromb Vasc Biol 2021; 41:883-897. [DOI: 10.1161/atvbaha.120.315133] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Objective:
Cardiovascular disease, a major cause of mortality and morbidity, exhibits sexual dimorphism since the onset of cardiovascular disease occurs later in women than in men. The loss of cardioprotection in older women may be due to an increase in arterial stiffness after menopause. Free fatty acid metabolites of polyunsaturated fatty acids, called oxylipins, are known to impact vessel function and may be responsible for the vascular benefits of polyunsaturated fatty acids. The objectives of this study were to compare the plasma oxylipin profiles of young females (20–55 years), older females (55
+
), and older males (55
+
) and to identify associations between oxylipins and cardiovascular disease risk factors, such as obesity and arterial stiffness.
Approach and Results:
We quantified plasma oxylipins by high-performance liquid chromatography–tandem mass spectrometry in archived samples taken from completed clinical trials. We identified 3 major 12-lipoxygenase products, 12-hydroxy-eicosatetraenoic acid, 12-hydroxy-eicosapentaenoic acid, and 14-hydroxy-docosahexaenoic acid, that are present at high levels in young females compared with older females and males. These oxylipins also decreased with obesity and displayed robust negative associations with arterial stiffness as assessed by brachial-ankle pulse wave velocity. According to multiple linear regression modeling, these associations were maintained even after correcting for body mass index category combined with either age, menopausal status, or estradiol levels. Using linear discriminant analysis, the combination of these 3 oxylipins effectively distinguished participants according to both brachial-ankle pulse wave velocity risk group and age.
Conclusions:
Higher 12-lipoxygenase oxylipin plasma concentrations associated with lower arterial stiffness in premenopausal females may be an important contributing factor to sex differences in cardiovascular disease.
Registration:
URL:
https://www.clinicaltrials.gov
; Unique identifiers: NCT01661543, NCT01562171, NCT01890330, NCT02571114 and NCT02317588.
Collapse
Affiliation(s)
- Samantha D. Pauls
- Department of Food and Human Nutritional Sciences (S.D.P., T.W., H.M.A., C.G.T., P.Z.), University of Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
| | - Youjia Du
- Department of Physiology and Pathophysiology (Y.D., C.G.T., P.Z.), University of Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
| | - Luc Clair
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
- Department of Economics, University of Winnipeg, Canada (L.C.)
| | - Tanja Winter
- Department of Food and Human Nutritional Sciences (S.D.P., T.W., H.M.A., C.G.T., P.Z.), University of Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
| | - Harold M. Aukema
- Department of Food and Human Nutritional Sciences (S.D.P., T.W., H.M.A., C.G.T., P.Z.), University of Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
| | - Carla G. Taylor
- Department of Food and Human Nutritional Sciences (S.D.P., T.W., H.M.A., C.G.T., P.Z.), University of Manitoba, Canada
- Department of Physiology and Pathophysiology (Y.D., C.G.T., P.Z.), University of Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
| | - Peter Zahradka
- Department of Food and Human Nutritional Sciences (S.D.P., T.W., H.M.A., C.G.T., P.Z.), University of Manitoba, Canada
- Department of Physiology and Pathophysiology (Y.D., C.G.T., P.Z.), University of Manitoba, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, Winnipeg, Canada (S.D.P., Y.D., L.C., T.W., H.M.A., C.G.T., P.Z.)
| |
Collapse
|
21
|
Moh MC, Low S, Ng TP, Ang SF, Ang K, Sum CF, Subramaniam T, Lim SC. Association between depressive symptoms and pulse wave velocity is mediated by increased adiposity in older adults with type 2 diabetes. J Psychiatry Neurosci 2021; 46:E176-E183. [PMID: 33464781 PMCID: PMC7955836 DOI: 10.1503/jpn.200080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
BACKGROUND Studies investigating the association between depression and aortic stiffness in older patients with type 2 diabetes are lacking. We postulated an association between depressive symptoms and aortic stiffness, and this relationship may be mediated by increased adiposity. METHODS We analyzed participants with type 2 diabetes aged 55 years or older (n = 958). We measured aortic stiffness using carotid-femoral pulse wave velocity (cut-off ≥ 12 m/s) using the tonometry method. We defined depressive symptoms as a score of greater than 5 on the Geriatric Depression Scale-15 (GDS-15). Adiposity indices we assessed were body mass index, waist circumference, waistto-height ratio, visceral fat area and fat mass. RESULTS Among the participants, 27.2% had aortic stiffness, of whom 6.5% had depressive symptoms. Score on the GDS-15 was correlated with pulse wave velocity, and both variables were correlated with the adiposity markers we analyzed (all p < 0.05). Depressive symptoms were associated with pulse wave velocity (B = 1.79, 95% confidence interval [CI] 0.83-2.75) or aortic stiffness (risk ratio 1.60, 95% CI 1.10-2.33) in the unadjusted model. The association persisted after controlling for demographics, duration of diabetes, glycated hemoglobin, comorbidities and medications. Further adjustment for visceral fat area and fat mass in separate models reduced the association between depressive symptoms and pulse wave velocity or aortic stiffness. Mediation models revealed that the mediation proportions of fat mass and visceral fat area on the association between depressive symptoms and pulse wave velocity were 11.8% and 9.7%, respectively. A preliminary analysis of longitudinal data (n = 184) showed similar findings. LIMITATIONS Causality cannot be inferred from the associations we observed. CONCLUSION Depressive symptoms are associated with elevated pulse wave velocity in older people with type 2 diabetes, and this relationship may be partially mediated by increased adiposity.
Collapse
Affiliation(s)
- Mei Chung Moh
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Serena Low
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Tze Pin Ng
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Su Fen Ang
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Keven Ang
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Chee Fang Sum
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Tavintharan Subramaniam
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| | - Su Chi Lim
- From the Clinical Research Unit, Khoo Teck Puat Hospital, Singapore (Moh, Low, S. Ang, K. Ang, Subramaniam, Lim); the Diabetes Centre, Admiralty Medical Centre, Khoo Teck Puat Hospital, Singapore (Low, Sum, Subramaniam, Lim); the Gerontology Research Programme, Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore (Ng); and the Saw Swee Hock School of Public Health, National University Hospital, Singapore (Lim)
| |
Collapse
|
22
|
Okuno K, Cicalese S, Elliott KJ, Kawai T, Hashimoto T, Eguchi S. Targeting Molecular Mechanism of Vascular Smooth Muscle Senescence Induced by Angiotensin II, A Potential Therapy via Senolytics and Senomorphics. Int J Mol Sci 2020; 21:ijms21186579. [PMID: 32916794 PMCID: PMC7555774 DOI: 10.3390/ijms21186579] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/03/2020] [Accepted: 09/07/2020] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is a prevalent issue in the global aging population. Premature vascular aging such as elevated arterial stiffness appears to be a major risk factor for CVD. Vascular smooth muscle cells (VSMCs) are one of the essential parts of arterial pathology and prone to stress-induced senescence. The pervasiveness of senescent VSMCs in the vasculature increases with age and can be further expedited by various stressing events such as oxidative stress, mitochondria dysfunction, endoplasmic reticulum stress, and chronic inflammation. Angiotensin II (AngII) can induce many of these responses in VSMCs and is thus considered a key regulator of VSMC senescence associated with CVD. Understanding the precise mechanisms and consequences of senescent cell accumulation may uncover a new generation of therapies including senolytic and senomorphic compounds against CVD. Accordingly, in this review article, we discuss potential molecular mechanisms of VSMC senescence such as those induced by AngII and the therapeutic manipulations of senescence to control age-related CVD and associated conditions such as by senolytic.
Collapse
Affiliation(s)
- Keisuke Okuno
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Stephanie Cicalese
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Katherine J. Elliott
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Tatsuo Kawai
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
| | - Tomoki Hashimoto
- Department of Neurosurgery and Neurobiology, Barrow Aneurysm and AVM Research Center, Barrow Neurological Institute, Phoenix, AZ 85013, USA;
| | - Satoru Eguchi
- Cardiovascular Research Center, Lewis Katz School of Medicine at Temple University, 3500 N. Broad Street, Philadelphia, PA 19140, USA; (K.O.); (S.C.); (K.J.E.); (T.K.)
- Correspondence:
| |
Collapse
|
23
|
Roy-Vallejo E, Galván-Román JM, Moldenhauer F, Real de Asúa D. Adults with Down syndrome challenge another paradigm: When aging no longer entails arterial hypertension. J Clin Hypertens (Greenwich) 2020; 22:1127-1133. [PMID: 32644285 DOI: 10.1111/jch.13930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 05/09/2020] [Accepted: 05/17/2020] [Indexed: 12/12/2022]
Abstract
The paradigmatic relationship between aging and atherosclerotic cardiovascular events does not apply to all patient populations. Though trisomy 21 (T21) and its phenotypic expression, Down syndrome (DS), are conditions that involve premature aging, the cardiovascular system of adults with DS appears to be particularly spared from this early senescence. Despite a higher prevalence of some classic cardiovascular risk factors in adults with DS than in the general population, such as dyslipidemia, obesity, or sedentarism, these individuals do not develop hypertension or suffer major cardiovascular events as they age. The protective factors that prevent the development of hypertension in T21 are not well established. Genes like RCAN1 and DYRK1A, both on chromosome 21 and over-expressed in adults with DS, appear to play a major role in cardiovascular prevention. Their regulation of the renin-angiotensin-aldosterone system (RAAS) and neprilysin synthesis could underlie the constitutive protection against arterial hypertension in adults with DS and explain the absence of increased arterial stiffness in this population. A better understanding of these molecular pathways could have enormous implications for the clinical management of adults with DS and might foster the development of novel therapeutic targets in cardiovascular prevention for the general population.
Collapse
Affiliation(s)
- Emilia Roy-Vallejo
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - José María Galván-Román
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - Fernando Moldenhauer
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| | - Diego Real de Asúa
- Adult Down Syndrome Unit, Department of Internal Medicine, Hospital Universitario de La Princesa, Madrid, Spain
| |
Collapse
|
24
|
张 艺, 盛 帅, 梁 庆, 张 莉. [Olmesartan inhibits age-associated migration and invasion of human aortic vascular smooth muscle cells by upregulating miR-3133 axis]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:499-505. [PMID: 32895132 PMCID: PMC7225100 DOI: 10.12122/j.issn.1673-4254.2020.04.08] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Indexed: 12/08/2022]
Abstract
OBJECTIVE To explore the effects of olmesartan on age-associated migration and invasion capacities and microRNA (miRAN) axis in human aortic vascular smooth muscle cells (HA-VSMCs). METHODS Cultured HA-VSMCs were divided into control group, bleomycin-mediated senescence (BLM) group and bleomycin + olmesartan treatment group. Wound-healing assay and Boyden chambers invasion assay were used to assess the changes in migration and invasion of the cells, gelatin zymography was used to analyze matrix metalloproteinase-2 (MMP-2) activation in the cells. The differentially expressed miRNAs were identified by miRNA microarray assay and validated by quantitative real-time PCR. MiR-3133 inhibitor was used to examine the effects of molecular manipulation of olmesartan on age-associated migration and invasion and MMP-2 activation in the cells. RESULTS Compared with those of the control group, the percentage of the repopulated cells and the number of cells crossing the basement membrane increased significantly in BLM group [(78.43±12.76)% vs (42.47±7.22)%, P < 0.05; 33.33±5.51 vs 13.00±4.36, P < 0.05]. A significant increase of MMP-2 activation was found in BLM group as compared with the control group (1.66 ± 0.27 vs 0.87 ± 0.13, P < 0.05). Olmesartan significantly inhibited BLM-induced enhancement of cell migration and invasion and MMP-2 secretion in the cells. MiR-3133 was significantly downregulated in BLM group and upregulated in olmesartan group. Transfection with miR-3133 inhibitor significantly reversed the effects of olmesartan on age-associated migration and invasion of the cells [(85.87±7.39)% vs (49.77±3.05)%; 34.67±2.31 vs 20.00±4.58, P < 0.05] and MMP-2 activation in the cells (1.76±0.19 vs 0.94±0.10, P < 0.05). CONCLUSIONS Olmesartan inhibits the migration and invasion of ageassociated HA-VSMCs probably by upregulating of the miR-3133 axis.
Collapse
Affiliation(s)
- 艺 张
- />广东药科大学附属第一医院心血管内科,广东 广州 510080Department of Cardiology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - 帅 盛
- />广东药科大学附属第一医院心血管内科,广东 广州 510080Department of Cardiology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - 庆阳 梁
- />广东药科大学附属第一医院心血管内科,广东 广州 510080Department of Cardiology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| | - 莉 张
- />广东药科大学附属第一医院心血管内科,广东 广州 510080Department of Cardiology, First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou 510080, China
| |
Collapse
|
25
|
Fhayli W, Boëté Q, Harki O, Briançon-Marjollet A, Jacob MP, Faury G. Rise and fall of elastic fibers from development to aging. Consequences on arterial structure-function and therapeutical perspectives. Matrix Biol 2019; 84:41-56. [PMID: 31493460 DOI: 10.1016/j.matbio.2019.08.005] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Revised: 08/03/2019] [Accepted: 08/26/2019] [Indexed: 12/12/2022]
Abstract
In the arteries of vertebrates, evolution has given rise to resilient macromolecular structures, elastin and elastic fibers, capable of sustaining an elevated blood pressure and smoothening the discontinuous blood flow and pressure generated by the heart. Elastic fibers are produced only during development and childhood, before being progressively degraded by mechanical stress and enzymatic activities during adulthood and aging. During this period, arterial elastic fiber calcification and loading of lipids also occur, all of these events conducting to arteriosclerosis. This leads to a progressive dysfunction of the large elastic arteries inducing elevated blood pressure as well as altered hemodynamics and organ perfusion, which induce more global malfunctions of the body during normal aging. Additionally, some arterial conditions occur more frequently with advancing age, such as atherosclerosis or aneurysms, which are called age-related diseases or pathological aging. The physiological or pathological degradation of elastic fibers and function of elastic arteries seemed to be rather inevitable over time. However, during the recent years, different molecules - including several ATP-dependent potassium channel openers, such as minoxidil - have been shown to re-induce elastin production and elastic fiber assembly, leading to improvements in the arterial structure and function or in organ perfusion. This review summarizes the changes in the arterial elastic fibers and structure from development until aging, and presents some of the potential pharmacotherapies leading to elastic fiber neosynthesis and arterial function improvement.
Collapse
Affiliation(s)
- Wassim Fhayli
- Univ. Grenoble Alpes, Inserm U1042, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - Quentin Boëté
- Univ. Grenoble Alpes, Inserm U1042, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | - Olfa Harki
- Univ. Grenoble Alpes, Inserm U1042, CHU Grenoble Alpes, HP2, 38000 Grenoble, France
| | | | - Marie-Paule Jacob
- INSERM, U1148, and Hopital Bichat-Claude Bernard, 46 rue Henri Huchard, 75877 Paris, France
| | - Gilles Faury
- Univ. Grenoble Alpes, Inserm U1042, CHU Grenoble Alpes, HP2, 38000 Grenoble, France.
| |
Collapse
|
26
|
Colpani O, Spinetti G. MicroRNAs orchestrating senescence of endothelial and vascular smooth muscle cells. ACTA ACUST UNITED AC 2019; 1:H75-H81. [PMID: 32923957 PMCID: PMC7439843 DOI: 10.1530/vb-19-0017] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 08/12/2019] [Indexed: 12/20/2022]
Abstract
During organism aging, the process of cellular senescence is triggered by critical stressors such as DNA damage, oncogenes, oxidative stress, and telomere erosion, and vascular cells are not exempted. Senescent cells stop proliferating but remain metabolically active producing pro-inflammatory signals in the environment collectively named senescence-associated secretory phenotype (SASP) that contribute to the amplification of the response to the neighbor and distant cells. Although the shift toward senescence is protective against tumors and needed during wound healing, the accumulation of senescent cells during aging due to an impairment of the immune system deputed to their clearance, can predispose to diseases of the cardiovascular system such as atherosclerosis. In this short review, we describe the main features of senescence of endothelial and smooth muscle cells and focus on the role non-coding RNAs of the microRNAs class in controlling this process. Finally, we discuss the potential of new strategies based on senescence removal in counteracting vascular disease burden.
Collapse
|