1
|
Cao Y, Huang M, Mao C, Wang X, Xu Y, Qian X, Ma C, Qiu W, Zhu Y. Structural changes in cerebral microvasculature induced by ferroptosis contribute to blood-brain barrier disruption in Alzheimer's disease: an autopsy study. Alzheimers Dement 2025; 21:e70103. [PMID: 40189804 PMCID: PMC11972978 DOI: 10.1002/alz.70103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/08/2025] [Accepted: 02/23/2025] [Indexed: 04/10/2025]
Abstract
INTRODUCTION Cerebrovascular lesions are associated with cognitive impairment. However, the impact of AD neuropathological changes (ADNC) on cerebral microvasculature is not completely understood. METHODS Twelve decedents with ADNC and 15 matched controls were selected from the Brain Bank. The changes in the median tunica and basement membrane-related extracellular matrix (ECM) contents of the microvasculature were quantified and compared. Additionally, we explored the related mechanisms of agrin in pericytes. RESULTS Venular collagenosis was significantly more severe in AD patients (p < 0.001), and ECM remolding was significantly correlated with ADNC. In the AD group, blood-brain barrier (BBB) disruption and decreased pericytes were observed. Finally, we confirmed that agrin induced ferroptosis in pericytes and BBB disruption in vitro. DISCUSSION Our data indicate that venular collagenosis and significant ECM remolding are important contributors to ADNC. The mechanism by which agrin's role in disrupting the BBB by inducing ferroptosis presents a potential new target. HIGHLIGHTS Changes in the median tunica and basement membrane-related ECM contents of the microvasculature were quantified in human brains. Venular collagenosis was significantly more severe in AD patients. In the AD group, BBB disruption and ECM remodeling were important contributors to AD neuropathological changes. Agrin disrupted the BBB by inducing ferroptosis in pericytes, which presents a potential new target.
Collapse
Affiliation(s)
- Yuan Cao
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Mei‐Ying Huang
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Chen‐Hui Mao
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Xue Wang
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterChinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Yuan‐Yuan Xu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterChinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Xiao‐Jing Qian
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterChinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Chao Ma
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterChinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Wen‐Ying Qiu
- Department of Human AnatomyHistology and EmbryologyInstitute of Basic Medical SciencesNeuroscience CenterChinese Academy of Medical SciencesSchool of Basic MedicinePeking Union Medical CollegeBeijingChina
| | - Yi‐Cheng Zhu
- Department of NeurologyState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
2
|
Kasal DA, Sena V, Huguenin GVB, De Lorenzo A, Tibirica E. Microvascular endothelial dysfunction in vascular senescence and disease. Front Cardiovasc Med 2025; 12:1505516. [PMID: 40041173 PMCID: PMC11878104 DOI: 10.3389/fcvm.2025.1505516] [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: 10/03/2024] [Accepted: 01/21/2025] [Indexed: 03/06/2025] Open
Abstract
Cardiovascular disease (CVD) is the main cause of morbidity and mortality in the adult and the elderly, with increasing prevalence worldwide. A growing body of research has focused on the earliest stage of vascular decline-endothelial dysfunction (ED)-which at the microvascular level can anticipate in decades the diagnosis of CVD. This review aims to provide a prospect of the literature regarding the development of ED as an indissociable feature of the aging of the cardiovascular system, highlighting the role of inflammation in the process. Vascular aging consists of a lifelong continuum, which starts with cell respiration and its inherent production of reactive oxygen species. Molecular imbalance is followed by cellular epigenetic changes, which modulate immune cells, such as macrophage and lymphocyte subtypes. These mechanisms are influenced by lifestyle habits, which affect inflammation hotspots in organism, such as visceral fat and gut microbiota. The process can ultimately lead to an environment committed to the loss of the physiological functions of endothelial cells. In addition, we discuss lifestyle changes targeting the connection between age-related inflammation and vascular dysfunction. Addressing microvascular ED represents a critical endeavor in order to prevent or delay vascular aging and associated diseases.
Collapse
Affiliation(s)
- Daniel A. Kasal
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
- Internal Medicine Department, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Viviane Sena
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Grazielle Vilas Bôas Huguenin
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
- Nutrition and Dietetics Department, Fluminense Federal University, Rio de Janeiro, Brazil
| | - Andrea De Lorenzo
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
| | - Eduardo Tibirica
- Research and Teaching Department, National Institute of Cardiology, Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Fu X, Zhao Y, Cui X, Huang S, Lv Y, Li C, Gong F, Yang Z, Yang X, Xiao R. Cxcl9 modulates aging associated microvascular metabolic and angiogenic dysfunctions in subcutaneous adipose tissue. Angiogenesis 2025; 28:17. [PMID: 39934436 PMCID: PMC11813824 DOI: 10.1007/s10456-025-09970-y] [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: 10/29/2024] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
Microvascular aging, predominantly driven by endothelial cells (ECs) dysfunction, is a critical early event in cardiovascular diseases. However, the specific effects of aging on ECs across the microvascular network segments and the associated mechanisms are not fully understood. In this study, we detected a microvascular rarefaction and a decreased proportion of venular ECs in the subcutaneous adipose tissue of aged mice using light-sheet immunofluorescence microscopy and single-cell RNA sequencing. Moreover, aged ECs, especially in the venular subtype, exhibited a pseudotemporal transition to a terminal state characterized by diminished oxidative phosphorylation and strengthened cytokine signaling. Metabolic flux balance analysis predicted that among the 13 differentially expressed cytokines identified in aged EC subpopulations, Cxcl9 was strongly correlated with impaired oxidative phosphorylation in aged ECs. It was further validated using microvascular ECs treated with Cxcl9. Notably, the G protein-coupled receptor signaling pathway was subsequently suppressed, in which Aplnr suppression was also observed in aged ECs, contributing to their impaired energy metabolism and reduced angiogenesis. Based on these findings, we propose Cxcl9 as a biomarker for aging-related dysfunction of microvascular ECs, suggesting that targeting Cxcl9 signaling may help combat microvascular aging.
Collapse
Affiliation(s)
- Xin Fu
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Yu Zhao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Xiwei Cui
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Siyuan Huang
- Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, P. R. China
| | - Yanze Lv
- Department of Hemangioma and Vascular Malformation of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
| | - Chen Li
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Fuxing Gong
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Zhigang Yang
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China
| | - Xiaonan Yang
- Department of Hemangioma and Vascular Malformation of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China
| | - Ran Xiao
- Research Center of Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, P. R. China.
- Key Laboratory of Tissue and Organ Regeneration, Chinese Academy of Medical Sciences, Beijing, P. R. China.
| |
Collapse
|
4
|
Wright SA, Lennon R, Greenhalgh AD. Basement membranes' role in immune cell recruitment to the central nervous system. J Inflamm (Lond) 2024; 21:53. [PMID: 39707430 DOI: 10.1186/s12950-024-00426-6] [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: 09/26/2024] [Accepted: 12/10/2024] [Indexed: 12/23/2024] Open
Abstract
Basement membranes form part of the extracellular matrix (ECM), which is the structural basis for all tissue. Basement membranes are cell-adherent sheets found between cells and vascular endothelia, including those of the central nervous system (CNS). There is exceptional regional specialisation of these structures, both in tissue organisation and regulation of tissue-specific cellular processes. Due to their location, basement membranes perform a key role in immune cell trafficking and therefore are important in inflammatory processes causing or resulting from CNS disease and injury. This review will describe basement membranes in detail, with special focus on the brain. We will cover how genetic changes drive brain pathology, describe basement membranes' role in immune cell recruitment and how they respond to various brain diseases. Understanding how basement membranes form the junction between the immune and central nervous systems will be a major advance in understanding brain disease.
Collapse
Affiliation(s)
- Shaun A Wright
- Lydia Becker Institute of Immunology and Inflammation, Division, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Rachel Lennon
- Cell Matrix Biology & Regenerative Medicine and Wellcome Centre for Cell-Matrix Research, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - Andrew D Greenhalgh
- Lydia Becker Institute of Immunology and Inflammation, Division, Division of Immunology, Immunity to Infection and Respiratory Medicine, School of Biological Science, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK.
- The University of Manchester, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
5
|
Alvares TS, Maturana FM, Soares RN. Sex differences in the predictors of skeletal muscle microvascular reactivity in older individuals. Maturitas 2024; 189:108115. [PMID: 39276434 DOI: 10.1016/j.maturitas.2024.108115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/17/2024] [Accepted: 09/05/2024] [Indexed: 09/17/2024]
Abstract
Aging is associated with an increased risk of cardiovascular disease and vascular dysfunction. Reduced nitric oxide bioavailability is considered one of the key mechanisms underlying vascular dysfunction in large arteries of older adults. However, the relationship between cardiovascular disease risk factors, nitric oxide bioavailability, and skeletal muscle microvascular reactivity, an early hallmark in cardiovascular disease progression, is unclear in older individuals. Also uncertain is whether this relationship is influenced by sex. Therefore, this study assessed the association between cardiovascular disease risk factors, circulating markers of nitric oxide availability (plasma nitrate and nitrite), and skeletal muscle microvascular reactivity in older individuals. First, we confirmed in a cohort of young and older individuals that aging is associated with skeletal muscle microvascular dysfunction. Next, we observed that skeletal muscle microvascular reactivity (P = 0.653; η2 = 0.016) and circulating nitric oxide metabolites (Nitrate: P = 0.641, η2 = 0.011; Nitrite: P = 0.560, η2 = 0.017; NOx: P = 0.639, η2 = 0.011) did not differ between older males and females. Finally, using multivariate regression models, we found that: (i) the number of cardiovascular risk factors was negatively associated with skeletal muscle microvascular reactivity in older males and females (B = -0.132, P = 0.044); (ii) the relationship between plasma nitrite and skeletal muscle microvascular reactivity was influenced by sex (F = 6.837, P = 0.016); and (iii) skeletal muscle microvascular reactivity in older females displayed a strong positive association with plasma nitrite (R2 = 0.720, P < 0.001). While the impact of cardiovascular disease risk factors on skeletal muscle microvascular reactivity was not influenced by sex, sex-related discrepancies were found in the relationship between nitric oxide bioavailability and skeletal muscle microvascular reactivity in older individuals.
Collapse
Affiliation(s)
- Thiago Silveira Alvares
- Multidisciplinary Center UFRJ-Macaé, Federal University of Rio de Janeiro, Macaé, Rio de Janeiro, Brazil.
| | | | | |
Collapse
|
6
|
Ahmed B, Rahman AA, Lee S, Malhotra R. The Implications of Aging on Vascular Health. Int J Mol Sci 2024; 25:11188. [PMID: 39456971 PMCID: PMC11508873 DOI: 10.3390/ijms252011188] [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: 09/18/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
Vascular aging encompasses structural and functional changes in the vasculature, significantly contributing to cardiovascular diseases, which are the leading cause of death globally. The incidence and prevalence of these diseases increase with age, with most morbidity and mortality attributed to myocardial infarction and stroke. Diagnosing and intervening in vascular aging while understanding the mechanisms behind age-induced vascular phenotypic and pathophysiological alterations offers the potential for delaying and preventing cardiovascular mortality in an aging population. This review delves into various aspects of vascular aging by examining age-related changes in arterial health at the cellular level, including endothelial dysfunction, cellular senescence, and vascular smooth muscle cell transdifferentiation, as well as at the structural level, including arterial stiffness and changes in wall thickness and diameter. We also explore aging-related changes in perivascular adipose tissue deposition, arterial collateralization, and calcification, providing insights into the physiological and pathological implications. Overall, aging induces phenotypic changes that augment the vascular system's susceptibility to disease, even in the absence of traditional risk factors, such as hypertension, diabetes, obesity, and smoking. Overall, age-related modifications in cellular phenotype and molecular homeostasis increase the vulnerability of the arterial vasculature to structural and functional alterations, thereby accelerating cardiovascular risk. Increasing our understanding of these modifications is crucial for success in delaying or preventing cardiovascular diseases. Non-invasive techniques, such as measuring carotid intima-media thickness, pulse wave velocity, and flow-mediated dilation, as well as detecting vascular calcifications, can be used for the early detection of vascular aging. Targeting specific pathological mechanisms, such as cellular senescence and enhancing angiogenesis, holds promise for innovative therapeutic approaches.
Collapse
Affiliation(s)
- Bulbul Ahmed
- Evans Department of Medicine and Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA 02118, USA;
| | - Ahmed A. Rahman
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Sujin Lee
- Division of Vascular Surgery, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA;
| | - Rajeev Malhotra
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
7
|
Myburgh-Jacobsz CE, Botha-Le Roux S, Kotliar K, Wentzel A, Jacobs A, De Boever P, Goswami N, Strijdom H, Smith W. Retinal Vessel Functional Responses in South Africans Living With and Without HIV: The EndoAfrica-NWU Study. Microcirculation 2024; 31:e12878. [PMID: 39106121 DOI: 10.1111/micc.12878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2023] [Revised: 07/05/2024] [Accepted: 07/17/2024] [Indexed: 08/09/2024]
Abstract
OBJECTIVES The effects of HIV and antiretroviral therapy (ART) on microvascular function are poorly explored. We compared retinal vessel functional responses to flicker light-induced provocation (FLIP) in people living with HIV (PLWH) and people living without HIV (PLWoutH). METHODS We included 115 PLWH and 51 PLWoutH with a median age of 41 years. Treated PLWH received similar first-line fixed-dose combination ART. Clinical characteristics and retinal vessels functional responses to FLIP were compared in (a) PLWH and PLWoutH; and (b) PLWH groups stratified by the median of (i) CD4-count (511 cells/mm3), (ii) viral load (50 copies/mL), and (iii) ART duration (57.6 months). RESULTS PLWH were older, smoked more, and had a lower prevalence of hypertension than PLWoutH (p < 0.05). Almost 64% of PLWH were infected for more than 5 years. Retinal vessel responses to FLIP were similar between PLWH and PLWoutH after taking confounders into account. In addition, PLWH subgroups stratified according to immuno-virological status by CD4-count, viral load, and ART duration showed no differences in retinal vessel responses to FLIP. CONCLUSION Living with HIV and receiving ART were not associated with altered microvascular function as assessed with dynamic retinal vessel analysis in a South African case-control study.
Collapse
Affiliation(s)
| | - Shani Botha-Le Roux
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Konstantin Kotliar
- Department of Medical Engineering and Technomathematics, Aachen University of Applied Sciences, Juelich, Germany
| | - Annemarie Wentzel
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Adriaan Jacobs
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| | - Patrick De Boever
- Centre for Environmental Sciences, Hasselt University, Diepenbeek, Belgium
- Antwerp University Hospital (UZA), Edegem, Belgium
| | - Nandu Goswami
- Gravitational Physiology and Medicine Research Unit, Division of Physiology and Pathophysiology, Medical University of Graz, Graz, Austria
- Center for Space and Aviation Health, College of Medicine, Mohammed bin Rashid University of Medicine and Health Sciences, Dubai, UAE
| | - Hans Strijdom
- Centre for Cardio-Metabolic Research in Africa, Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Wayne Smith
- Hypertension in Africa Research Team (HART), Faculty of Health Sciences, North-West University, Potchefstroom, South Africa
- MRC Research Unit for Hypertension and Cardiovascular Disease, North-West University, Potchefstroom, South Africa
| |
Collapse
|
8
|
Dimitriadis K, Pyrpyris N, Sakalidis A, Dri E, Iliakis P, Tsioufis P, Tatakis F, Beneki E, Fragkoulis C, Aznaouridis K, Tsioufis K. ANOCA updated: From pathophysiology to modern clinical practice. CARDIOVASCULAR REVASCULARIZATION MEDICINE 2024:S1553-8389(24)00672-9. [PMID: 39341735 DOI: 10.1016/j.carrev.2024.09.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/03/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024]
Abstract
Lately, a large number of stable ischemic patients, with no obstructed coronary arteries are being diagnosed. Despite this condition, which is being described as angina with no obstructive coronary arteries (ANOCA), was thought to be benign, recent evidence report that it is associated with increased risk for adverse cardiovascular outcomes. ANOCA is more frequent in women and, pathophysiologically, it is predominantly related with microvascular dysfunction, while other factors, such as endothelial dysfunction, inflammation and autonomic nervous system seem to also play a major role to its development, while other studies implicate ANOCA and microvascular dysfunction in the pathogenesis of heart failure with preserved ejection fraction. For establishing an ANOCA diagnosis, measurement including coronary flow reserve (CFR), microvascular resistance (IMR) and hyperemic microvascular resistance (HMR) are mostly used in clinical practice. In addition, new modalities, such as optical coherence tomography (OCT) are being tested and show promising results for future diagnostic use. Regarding management, pharmacotherapy consists of a wide selection of drugs, according to the respected pathophysiology of the disease (vasospastic angina or microvascular dysfunction), while research for new treatment options including interventional techniques, is currently ongoing. This review, therefore, aims to provide a comprehensive analysis of all aspects related to ANOCA, from pathophysiology to clinical managements, as well as clinical implications and suggestions for future research efforts, which will help advance our understanding of the syndrome and establish more, evidence-based, therapies.
Collapse
Affiliation(s)
- Kyriakos Dimitriadis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece.
| | - Nikolaos Pyrpyris
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Athanasios Sakalidis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Eirini Dri
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Panagiotis Iliakis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Panagiotis Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Fotis Tatakis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Eirini Beneki
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Christos Fragkoulis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Konstantinos Aznaouridis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| | - Konstantinos Tsioufis
- First Department of Cardiology, School of Medicine, National and Kapodistrian University of Athens, Hippokration Hospital, Athens, Greece
| |
Collapse
|
9
|
Joseph S, Woo J, Robbins CB, Haystead A, Stinnett S, Grewal DS, Fekrat S. Longitudinal Assessment of Peripapillary Microvasculature Using Optical Coherence Tomography Angiography in Cognitively Normal Adults. JOURNAL OF VITREORETINAL DISEASES 2024; 8:571-576. [PMID: 39318979 PMCID: PMC11418659 DOI: 10.1177/24741264241263167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/26/2024]
Abstract
Introduction: To evaluate longitudinal peripapillary changes in cognitively normal older adults using optical coherence tomography (OCT) and OCT angiography (OCTA). Methods: Participants older than 50 years with no history of neurodegenerative disease or cognitive impairment were prospectively enrolled. OCT and OCTA images were obtained at the first visit and 2 years later. Results: The study comprised 189 eyes of 111 adults with a mean age (±SD) of 69.3 ± 5.8 years and mean follow-up of 2.1 ± 0.5 years. Woman experienced slower rate of decline than men in capillary perfusion density (0.000% ± 0.005% vs -0.002% ± 0.004%; P = .038) and retinal nerve fiber layer (RNFL) thickness (0.133 ± 1.617 µm vs -0.659 ± 1.431 µm; P = .008). At both timepoints, after controlling for sex, the capillary perfusion density (P < .001), capillary flux index (P < .001), and RNFL thickness (P = .005) were lower in older participants. The mean capillary perfusion density was higher in women than in men at both timepoints (P = .01 and P = .002, respectively), with no significant differences in the capillary flux index and RNFL thickness. Conclusions: In cognitively normal adults, there is a significant reduction in peripapillary capillary perfusion density, the capillary flux index, and RNFL thickness associated with aging beyond 50 years. Women had higher capillary perfusion density values with slower rates of change in capillary perfusion density and RNFL thickness. These values can serve as benchmarks, and variations could be suspicious for a pathologic process.
Collapse
Affiliation(s)
- Suzanna Joseph
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Joshua Woo
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Cason B. Robbins
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Alice Haystead
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Sandra Stinnett
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Dilraj S. Grewal
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| | - Sharon Fekrat
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
- iMIND Research Group, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
10
|
Rezzani R, Favero G, Cominelli G, Pinto D, Rinaldi F. Skin Aging and the Upcoming Role of Ferroptosis in Geroscience. Int J Mol Sci 2024; 25:8238. [PMID: 39125810 PMCID: PMC11311626 DOI: 10.3390/ijms25158238] [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: 07/01/2024] [Revised: 07/25/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
The skin is considered the most important organ system in mammals, and as the population ages, it is important to consider skin aging and anti-aging therapeutic strategies. Exposure of the skin to various insults induces significant changes throughout our lives, differentiating the skin of a young adult from that of an older adult. These changes are caused by a combination of intrinsic and extrinsic aging. We report the interactions between skin aging and its metabolism, showing that the network is due to several factors. For example, iron is an important nutrient for humans, but its level increases with aging, inducing deleterious effects on cellular functions. Recently, it was discovered that ferroptosis, or iron-dependent cell death, is linked to aging and skin diseases. The pursuit of new molecular targets for ferroptosis has recently attracted attention. Prevention of ferroptosis is an effective therapeutic strategy for the treatment of diseases, especially in old age. However, the pathological and biological mechanisms underlying ferroptosis are still not fully understood, especially in skin diseases such as melanoma and autoimmune diseases. Only a few basic studies on regulated cell death exist, and the challenge is to turn the studies into clinical applications.
Collapse
Affiliation(s)
- Rita Rezzani
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.F.); (G.C.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy; (D.P.); (F.R.)
- Italian Society for the Study of Orofacial Pain (Società Italiana Studio Dolore Orofacciale—SISDO), 25123 Brescia, Italy
| | - Gaia Favero
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.F.); (G.C.)
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy; (D.P.); (F.R.)
| | - Giorgia Cominelli
- Anatomy and Physiopathology Division, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (G.F.); (G.C.)
| | - Daniela Pinto
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy; (D.P.); (F.R.)
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy
| | - Fabio Rinaldi
- Interdipartimental University Center of Research “Adaption and Regeneration of Tissues and Organs (ARTO)”, University of Brescia, 25123 Brescia, Italy; (D.P.); (F.R.)
- Human Microbiome Advanced Project Institute, 20129 Milan, Italy
| |
Collapse
|
11
|
Galante D, Migliaro S, Di Giusto F, Anastasia G, Petrolati E, Vicerè A, Zimbardo G, Cialdella P, Romagnoli E, Aurigemma C, Burzotta F, Trani C, Martin-Reyes R, Baptista SB, Faria D, Amabile N, Raposo L, Crea F, Leone AM. Age and Vasodilator Response to Different Hyperemic Agents: Adenosine versus Contrast Medium. Rev Cardiovasc Med 2024; 25:239. [PMID: 39139436 PMCID: PMC11317353 DOI: 10.31083/j.rcm2507239] [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: 12/19/2023] [Revised: 02/18/2024] [Accepted: 03/05/2024] [Indexed: 08/15/2024] Open
Abstract
Background Age-related remodelling has the potential to affect the microvascular response to hyperemic stimuli. However, its precise effects on the vasodilatory response to adenosine and contrast medium, as well as its influence on fractional flow reserve (FFR) and contrast fractional flow reserve (cFFR), have not been previously investigated. We investigate the impact of age on these indices. Methods We extrapolated data from the post-revascularization optimization and physiological evaluation of intermediate lesions using fractional flow reserve (PROPHET-FFR) and The Multi-center Evaluation of the Accuracy of the Contrast MEdium INduced Pd/Pa RaTiO in Predicting (MEMENTO) studies. Only lesions with a relevant vasodilatory response to adenosine and contrast medium were considered of interest. A total of 2080 patients, accounting for 2294 pressure recordings were available for analysis. The cohort was stratified into three age terciles. Age-dependent correlations with FFR, cFFR, distal pressure/aortic pressure (Pd/Pa) and instantaneous wave-free ratio (iFR) were calculated. The vasodilatory response was calculated in 1619 lesions (with both FFR and cFFR) as the difference between resting and hyperaemic pressure ratios and correlated with aging. The prevalence of FFR-cFFR discordance was assessed. Results Age correlated positively to FFR (r = 0.062, p = 0.006), but not with cFFR (r = 0.024, p = 0.298), Pd/Pa (r = -0.015, p = 0.481) and iFR (r = -0.026, p = 0.648). The hyperemic response to adenosine (r = -0.102, p ≤ 0.0001) and to contrast medium (r = -0.076, p = 0.0023) showed a negative correlation with age. When adjusted for potential confounders, adenosine induced hyperaemia was negatively associated with age (p = 0.04 vs p = 0.08 for cFFR). Discordance decreased across age terciles (14.64% vs 12.72% vs 10.12%, p = 0.032). Conclusions As compared to adenosine, contrast induced hyperaemia appeared to be less affected by age. cFFR may be considered a more stable and reproducible tool to assess epicardial stenosis in elderly patients. Clinical Trial Registration PROPHET-FFR STUDY, Clinicaltrials.gov (NCT05056662).
Collapse
Affiliation(s)
- Domenico Galante
- Diagnostic, Interventional and Acute Cardiac Care Unit, Ospedale Isola Tiberina – Gemelli Isola, 00186 Rome, Italy
| | - Stefano Migliaro
- Clinical, Interventional and Hemodynamic Cardiology Unit, Aurelia Hospital, 00165 Rome, Italy
| | - Federico Di Giusto
- Cardiology Unit, C. and G. Mazzoni Hospital, 63100 AST Ascoli Piceno, Italy
| | - Gianluca Anastasia
- Department of Internal Medicine, University of Genoa, 16132 Genova, Italy
| | - Edoardo Petrolati
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Vicerè
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giuseppe Zimbardo
- Cardiologia e Unità terapia intensiva, Policlinico Casilino, 00169 Rome, Italy
| | - Pio Cialdella
- Cardiologia e Unità terapia intensiva, Policlinico Casilino, 00169 Rome, Italy
| | - Enrico Romagnoli
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Cardiovascular Sciences Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Cristina Aurigemma
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Cardiovascular Sciences Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Francesco Burzotta
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Cardiovascular Sciences Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Carlo Trani
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Department of Cardiovascular Sciences Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy
| | - Roberto Martin-Reyes
- Unidad Integral de Cardiologia (UICAR). Hospital Universitario La Luz Quironsalud and Hospital Universitario Fundacion Jimenez Díaz Quironsalud, 28003 Madrid, Spain
| | - Sergio Bravo Baptista
- Cardiology Department, Hospital Prof. Doutor Fernando Fonseca, 2720-276 Amadora, Portugal
- Centro Cardiovascular da Universidade de Lisboa, Faculdade de Medicina da Universidade de Lisboa, 1649-028 Lisboa, Portugal
| | - Daniel Faria
- Cardiology Department, Hospital Prof. Doutor Fernando Fonseca, 2720-276 Amadora, Portugal
| | - Nicolas Amabile
- Cardiology Department, L’Institut Mutualiste Montsouris, 75014 Paris, France
| | - Luis Raposo
- Department of Cardiology, Centro Hospitalar de Lisboa Ocidental, 1300-598 Lisbon, Portugal
| | - Filippo Crea
- Diagnostic, Interventional and Acute Cardiac Care Unit, Ospedale Isola Tiberina – Gemelli Isola, 00186 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Maria Leone
- Diagnostic, Interventional and Acute Cardiac Care Unit, Ospedale Isola Tiberina – Gemelli Isola, 00186 Rome, Italy
- Department of Cardiovascular and Pneumological Sciences, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| |
Collapse
|
12
|
Hoek R, van Diemen PA, Raijmakers PG, Driessen RS, Somsen YBO, de Winter RW, Jukema RA, Twisk JWR, Robbers LFHJ, van der Harst P, Saraste A, Lubberink M, Sörensen J, Knaapen P, Knuuti J, Danad I. Determining Hemodynamically Significant Coronary Artery Disease: Patient-Specific Cutoffs in Quantitative Myocardial Blood Flow Using [ 15O]H 2O PET Imaging. J Nucl Med 2024; 65:1113-1121. [PMID: 38724275 DOI: 10.2967/jnumed.123.267195] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 04/08/2024] [Indexed: 07/03/2024] Open
Abstract
Currently, cutoffs of quantitative [15O]H2O PET to detect fractional flow reserve (FFR)-defined coronary artery disease (CAD) were derived from a single cohort that included patients without prior CAD. However, prior CAD, sex, and age can influence myocardial blood flow (MBF). Therefore, the present study determined the influence of prior CAD, sex, and age on optimal cutoffs of hyperemic MBF (hMBF) and coronary flow reserve (CFR) and evaluated whether cutoff optimization enhanced diagnostic performance of quantitative [15O]H2O PET against an FFR reference standard. Methods: Patients with chronic coronary symptoms underwent [15O]H2O PET and invasive coronary angiography with FFR. Optimal cutoffs for patients with and without prior CAD and subpopulations based on sex and age were determined. Results: This multicenter study included 560 patients. Optimal cutoffs were similar for patients with (n = 186) and without prior CAD (hMBF, 2.3 vs. 2.3 mL·min-1·g-1; CFR, 2.7 vs. 2.6). Females (n = 190) had higher hMBF cutoffs than males (2.8 vs. 2.3 mL·min-1·g-1), whereas CFRs were comparable (2.6 vs. 2.7). However, female sex-specific hMBF cutoff implementation decreased diagnostic accuracy as compared with the cutoff of 2.3 mL·min-1·g-1 (72% vs. 82%, P < 0.001). Patients aged more than 70 y (n = 79) had lower hMBF (1.7 mL·min-1·g-1) and CFR (2.3) cutoffs than did patients aged 50 y or less, 51-60 y, and 61-70 y (hMBF, 2.3-2.4 mL·min-1·g-1; CFR, 2.7). Age-specific cutoffs in patients aged more than 70 y yielded comparable accuracy to the previously established cutoffs (hMBF, 72% vs. 76%, P = 0.664; CFR, 80% vs. 75%, P = 0.289). Conclusion: Patients with and without prior CAD had similar [15O]H2O PET cutoffs for detecting FFR-defined significant CAD. Stratifying patients according to sex and age led to different optimal cutoffs; however, these values did not translate into an increased overall accuracy as compared with previously established thresholds for MBF.
Collapse
Affiliation(s)
- Roel Hoek
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
| | - Pepijn A van Diemen
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pieter G Raijmakers
- Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Roel S Driessen
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Yvemarie B O Somsen
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ruben W de Winter
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Ruurt A Jukema
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Jos W R Twisk
- Department of Epidemiology and Data Science, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Lourens F H J Robbers
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Pim van der Harst
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antti Saraste
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland; and
| | - Mark Lubberink
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Jens Sörensen
- Nuclear Medicine and PET, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Paul Knaapen
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Juhani Knuuti
- Turku PET Centre, Turku University Hospital and University of Turku, Turku, Finland
| | - Ibrahim Danad
- Department of Cardiology, Amsterdam University Medical Center, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands;
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
13
|
Huang BB, Fawzi AA. Hypertension Likely Drives Arteriolar Wall Thickening in Preclinical Diabetic Retinopathy While Diabetes Drives Wall Thickness in Clinical Retinopathy. Transl Vis Sci Technol 2024; 13:8. [PMID: 38874974 PMCID: PMC11182368 DOI: 10.1167/tvst.13.6.8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Accepted: 05/01/2024] [Indexed: 06/15/2024] Open
Abstract
Purpose Both hypertension and diabetes are known to increase the wall-to-lumen ratio (WLR) of retinal arterioles, but the differential effects are unknown. Here, we study the timing and relative impact of hypertension versus diabetes on the WLR in diabetic retinopathy (DR) to address this unresolved question. Methods This prospective cross-sectional study compared the retinal arteriolar WLR in 17 healthy eyes, 15 with diabetes but no apparent DR (DM no DR), and 8 with diabetic macular edema (DME) and either nonproliferative or proliferative DR. We imaged each arteriole using adaptive optics scanning laser ophthalmoscopy and measured the WLR using ImageJ. Multiple linear regression (MLR) was performed to estimate the effects of hypertension, diabetes, and age on the WLR. Results Both subjects with DM no DR and subjects with DME had significantly higher WLR than healthy subjects (0.36 ± 0.08 and 0.42 ± 0.08 vs. 0.29 ± 0.07, 1-way ANOVA P = 0.0009). MLR in healthy subjects and subjects with DM no DR showed hypertension had the strongest effect (regression coefficient = 0.08, P = 0.009), whereas age and diabetes were not significantly correlated with WLR. MLR in all three groups together (healthy, DM no DR, and DME) showed diabetes had the strongest effect (regression coefficient = 0.05, P = 0.02), whereas age and hypertension were not significantly correlated with WLR. Conclusions Hypertension may be an early driver of retinal arteriolar wall thickening in preclinical DR, independent of age or diabetes, whereas changes specific to DR may drive wall thickening in DME and later DR stages. Translational Relevance We offer a framework for understanding the relative contributions of hypertension and diabetes on the vascular wall, and emphasize the importance of hypertension control early in diabetes even before DR onset.
Collapse
Affiliation(s)
- Bonnie B. Huang
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Amani A. Fawzi
- Department of Ophthalmology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
14
|
Bullock-Palmer RP, Chareonthaitawee P, Fox E, Beache GM. Microvascular vasoregulatory dysfunction in African Americans - An enhanced opportunity for early prevention and treatment of atherosclerotic cardiovascular disease. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 40:100382. [PMID: 38586429 PMCID: PMC10994957 DOI: 10.1016/j.ahjo.2024.100382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024]
Abstract
Atherosclerotic cardiovascular disease and its risk factors and precursors are a major driver of disparities in cardiovascular health. This review examines reported evidence that vascular endothelial dysfunction, and its manifestation as coronary microvascular dysfunction, underlies observed excess morbidity and mortality in African Americans. Advanced imaging insights that reveal patho-mechanisms, along with population evidence from the Jackson Heart Study, and the growing evidence emanating from national and international clinical trials and registries are presented. We examine a physiological framework that recognizes insulin-resistant cardiometabolic underpinnings of the conditions of the American Heart Associations' Life's Essential Eight construct of cardiovascular health as a unifying basis that affords early prevention. Mechanistic-based therapeutic approaches, can subsequently be implemented to interrupt progression to adverse outcomes employing layered, or personalized, treatment strategies of a well-defined set of conditions or diseases. Remaining knowledge gaps are acknowledged.
Collapse
Affiliation(s)
- Renee P. Bullock-Palmer
- Department of Cardiology, Deborah Heart and Lung Center, Browns Mills NJ, Department of Medicine, Division of Cardiology, Thomas Jefferson University, Philadelphia, PA, United States of America
| | | | - Ervin Fox
- Division of Cardiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, United States of America
| | - Garth M. Beache
- Department of Radiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| |
Collapse
|
15
|
Wang P, Konja D, Singh S, Zhang B, Wang Y. Endothelial Senescence: From Macro- to Micro-Vasculature and Its Implications on Cardiovascular Health. Int J Mol Sci 2024; 25:1978. [PMID: 38396653 PMCID: PMC10889199 DOI: 10.3390/ijms25041978] [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: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/03/2024] [Indexed: 02/25/2024] Open
Abstract
Endothelial cells line at the most inner layer of blood vessels. They act to control hemostasis, arterial tone/reactivity, wound healing, tissue oxygen, and nutrient supply. With age, endothelial cells become senescent, characterized by reduced regeneration capacity, inflammation, and abnormal secretory profile. Endothelial senescence represents one of the earliest features of arterial ageing and contributes to many age-related diseases. Compared to those in arteries and veins, endothelial cells of the microcirculation exhibit a greater extent of heterogeneity. Microcirculatory endothelial senescence leads to a declined capillary density, reduced angiogenic potentials, decreased blood flow, impaired barrier properties, and hypoperfusion in a tissue or organ-dependent manner. The heterogeneous phenotypes of microvascular endothelial cells in a particular vascular bed and across different tissues remain largely unknown. Accordingly, the mechanisms underlying macro- and micro-vascular endothelial senescence vary in different pathophysiological conditions, thus offering specific target(s) for therapeutic development of senolytic drugs.
Collapse
Affiliation(s)
- Peichun Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Daniels Konja
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Sandeep Singh
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Beijia Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Yu Wang
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China; (P.W.); (D.K.); (S.S.); (B.Z.)
- Department of Pharmacology and Pharmacy, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
16
|
Zamboulis DE, Marr N, Lenzi L, Birch HL, Screen HRC, Clegg PD, Thorpe CT. The Interfascicular Matrix of Energy Storing Tendons Houses Heterogenous Cell Populations Disproportionately Affected by Aging. Aging Dis 2024; 15:295-310. [PMID: 37307816 PMCID: PMC10796100 DOI: 10.14336/ad.2023.0425-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/25/2023] [Indexed: 06/14/2023] Open
Abstract
Energy storing tendons such as the human Achilles and equine superficial digital flexor tendon (SDFT) are prone to injury, with incidence increasing with aging, peaking in the 5th decade of life in the human Achilles tendon. The interfascicular matrix (IFM), which binds tendon fascicles, plays a key role in energy storing tendon mechanics, and aging alterations to the IFM negatively impact tendon function. While the mechanical role of the IFM in tendon function is well-established, the biological role of IFM-resident cell populations remains to be elucidated. Therefore, the aim of this study was to identify IFM-resident cell populations and establish how these populations are affected by aging. Cells from young and old SDFTs were subjected to single cell RNA-sequencing, and immunolabelling for markers of each resulting population used to localise cell clusters. Eleven cell clusters were identified, including tenocytes, endothelial cells, mural cells, and immune cells. One tenocyte cluster localised to the fascicular matrix, whereas nine clusters localised to the IFM. Interfascicular tenocytes and mural cells were preferentially affected by aging, with differential expression of genes related to senescence, dysregulated proteostasis and inflammation. This is the first study to establish heterogeneity in IFM cell populations, and to identify age-related alterations specific to IFM-localised cells.
Collapse
Affiliation(s)
- Danae E. Zamboulis
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW1 0TU, UK.
| | - Neil Marr
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW1 0TU, UK.
| | - Luca Lenzi
- Centre for Genomic Research, University of Liverpool, Liverpool, L69 7ZB, UK.
| | - Helen L. Birch
- Department of Orthopaedics and Musculoskeletal Science, University College London, Royal National Orthopaedic Hospital, Stanmore, HA7 4LP, UK.
| | - Hazel R. C. Screen
- Institute of Bioengineering, School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS, UK.
| | - Peter D. Clegg
- Department of Musculoskeletal and AgingScience, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK.
| | - Chavaunne T. Thorpe
- Department of Comparative Biomedical Sciences, Royal Veterinary College, London, NW1 0TU, UK.
| |
Collapse
|
17
|
Yang YP, Chiu CT, Chao A, Yeh YC, Sun WZ, Liu CM, Chan WS. An observational study of microcirculation among healthy individuals by age and sex. Clin Hemorheol Microcirc 2024; 86:407-417. [PMID: 38073381 DOI: 10.3233/ch-231807] [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: 05/12/2024]
Abstract
OBJECTIVE This study measured normal ranges of microcirculatory parameters in healthy individuals and investigated differences in parameters by age and sex. METHODS Participants were enrolled into three groups with equal numbers of male and female: young (20-39 years), middle-aged (40-59 years), and elderly (60-79 years). Sublingual microcirculation images were obtained using the incident dark field (IDF). RESULTS A total of 75 female and 75 male healthy individuals were enrolled. The elderly group had a higher TVD (26.5 [2] vs. 25.2 [1.8]; p = 0.019) and a lower PPV (97 [2] vs. 98 [3]; p = 0.03) than did the young group. In the elderly group, systolic blood pressure (SBP) and mean arterial pressure (MAP) were moderately and positively correlated with MFI score (r = 0.407, p < 0.05, and r = 0.403, p < 0.05, respectively). The female participants had a lower MFI score than did the male participants (2.9 [2.8-3] vs. 3.0 [2.9-3]; p = 0.015). CONCLUSIONS This study revealed the range of microcirculatory parameters between different ages and sexes in healthy individuals. We found that blood pressure levels were correlated with microcirculatory parameters, especially in elders and female.
Collapse
Affiliation(s)
- Yun-Ping Yang
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Tang Chiu
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Anne Chao
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Yu-Chang Yeh
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wei-Zen Sun
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Min Liu
- Department of Anesthesiology, National Taiwan University Hospital, Taipei, Taiwan
| | - Wing-Sum Chan
- Department of Anesthesiology, Far Eastern Memorial Hospital, New Taipei, Taiwan
| |
Collapse
|
18
|
Cheng DCY, Climie RE, Shu M, Grieve SM, Kozor R, Figtree GA. Vascular aging and cardiovascular disease: pathophysiology and measurement in the coronary arteries. Front Cardiovasc Med 2023; 10:1206156. [PMID: 38089775 PMCID: PMC10715672 DOI: 10.3389/fcvm.2023.1206156] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 11/13/2023] [Indexed: 11/29/2024] Open
Abstract
Age is a key risk factor for cardiovascular disease, including atherosclerosis. However, pathophysiological disease processes in the arteries are not an inevitable feature of aging. Large cohort studies with arterial phenotyping along with clinical and demographic data are essential to better understand factors related to the susceptibility or resilience to age-related vascular pathophysiology in humans. This review explores the mechanisms by which vascular structure and function alters with age, and how these changes relate to cardiovascular pathophysiology and disease. Features of vascular aging in the coronary arteries have historically been difficult to quantify pre-mortem due to their size and location. However, non-invasive imaging modalities including CT Coronary Angiogram are now being used to assess coronary vascular age, and further advances in imaging analysis such as the CT Fat Attenuation Index will help provide further measurement of features associated with coronary vascular aging. Currently, markers of vascular aging are not used as therapeutic targets in routine clinical practice, but non-pharmacological interventions including aerobic exercise and low salt diet, as well as anti-hypertensives have been demonstrated to reduce arterial stiffness. Advances in imaging technology, both in acquisition and advanced analysis, as well as harmonisation of measurements for researchers across the globe will be invaluable in understanding what constitutes healthy vascular aging and in identifying features of vascular aging that are associated with coronary artery disease and its adverse outcomes. Assessing such images in large cohorts can facilitate improved definitions of resilient and susceptible phenotypes to vascular aging in the coronary arteries. This is a critical step in identifying further risk factors and biomarkers within these groups and driving forward the development of novel therapies aimed at slowing or stopping age-related vascular changes in the coronary arteries.
Collapse
Affiliation(s)
- Daniel C. Y. Cheng
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Rachel E. Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Matthew Shu
- Northern Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Stuart M. Grieve
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
| | - Rebecca Kozor
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
- Imaging and Phenotyping Laboratory, Charles Perkins Centre and Faculty of Medicine and Health, University of Sydney, Sydney, NSW, Australia
- Department of Cardiology, Royal North Shore Hospital, Sydney, NSW, Australia
| |
Collapse
|
19
|
Yee EM, Hauser CT, Petrocelli JJ, de Hart NMMP, Ferrara PJ, Bombyck P, Fennel ZJ, van Onselen L, Mookerjee S, Funai K, Symons JD, Drummond MJ. Treadmill training does not enhance skeletal muscle recovery following disuse atrophy in older male mice. Front Physiol 2023; 14:1263500. [PMID: 37942230 PMCID: PMC10628510 DOI: 10.3389/fphys.2023.1263500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/02/2023] [Indexed: 11/10/2023] Open
Abstract
Introduction: A hallmark of aging is poor muscle recovery following disuse atrophy. Efficacious strategies to enhance muscle recovery following disuse atrophy in aging are non-existent. Prior exercise training could result in favorable muscle morphological and cellular adaptations that may promote muscle recovery in aging. Here, we characterized the impact of exercise training on skeletal muscle inflammatory and metabolic profiles and cellular remodeling and function, together with femoral artery reactivity prior to and following recovery from disuse atrophy in aged male mice. We hypothesized that 12 weeks of treadmill training in aged male mice would improve skeletal muscle cellular remodeling at baseline and during recovery from disuse atrophy, resulting in improved muscle regrowth. Methods: Physical performance, ex vivo muscle and vascular function, tissue and organ mass, hindlimb muscle cellular remodeling (macrophage, satellite cell, capillary, myofiber size, and fibrosis), and proteolytic, inflammatory, and metabolic muscle transcripts were evaluated in aged exercise-trained and sedentary mice. Results: We found that at baseline following exercise training (vs. sedentary mice), exercise capacity and physical function increased, fat mass decreased, and endothelial function improved. However, exercise training did not alter tibialis anterior or gastrocnemius muscle transcriptional profile, macrophage, satellite cell, capillarity or collagen content, or myofiber size and only tended to increase tibialis mass during recovery from disuse atrophy. Conclusion: While exercise training in old male mice improved endothelial function, physical performance, and whole-body tissue composition as anticipated, 12 weeks of treadmill training had limited impact on skeletal muscle remodeling at baseline or in response to recovery following disuse atrophy.
Collapse
Affiliation(s)
- Elena M. Yee
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, United States
| | - Carson T. Hauser
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
| | - Jonathan J. Petrocelli
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, United States
| | - Naomi M. M. P. de Hart
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
| | - Patrick J. Ferrara
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, United States
| | - Princess Bombyck
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, United States
| | - Zachary J. Fennel
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| | - Lisha van Onselen
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, United States
| | - Sohom Mookerjee
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
| | - Katsuhiko Funai
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| | - J. David Symons
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| | - Micah J. Drummond
- Department of Physical Therapy and Athletic Training, University of Utah, Salt Lake City, UT, United States
- Department of Nutrition and Integrative Physiology, University of Utah, Salt Lake City, UT, United States
- Molecular Medicine Program, University of Utah, Salt Lake City, UT, United States
| |
Collapse
|
20
|
Owens CD, Bonin Pinto C, Mukli P, Szarvas Z, Peterfi A, Detwiler S, Olay L, Olson AL, Li G, Galvan V, Kirkpatrick AC, Balasubramanian P, Tarantini S, Csiszar A, Ungvari Z, Prodan CI, Yabluchanskiy A. Vascular mechanisms leading to progression of mild cognitive impairment to dementia after COVID-19: Protocol and methodology of a prospective longitudinal observational study. PLoS One 2023; 18:e0289508. [PMID: 37535668 PMCID: PMC10399897 DOI: 10.1371/journal.pone.0289508] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 07/19/2023] [Indexed: 08/05/2023] Open
Abstract
INTRODUCTION Mild cognitive impairment (MCI) is a prodromal stage to dementia, affecting up to 20% of the aging population worldwide. Patients with MCI have an annual conversion rate to dementia of 15-20%. Thus, conditions that increase the conversion from MCI to dementia are of the utmost public health concern. The COVID-19 pandemic poses a significant impact on our aging population with cognitive decline as one of the leading complications following recovery from acute infection. Recent findings suggest that COVID-19 increases the conversion rate from MCI to dementia in older adults. Hence, we aim to uncover a mechanism for COVID-19 induced cognitive impairment and progression to dementia to pave the way for future therapeutic targets that may mitigate COVID-19 induced cognitive decline. METHODOLOGY A prospective longitudinal study is conducted at the University of Oklahoma Health Sciences Center. Patients are screened in the Department of Neurology and must have a formal diagnosis of MCI, and MRI imaging prior to study enrollment. Patients who meet the inclusion criteria are enrolled and followed-up at 18-months after their first visit. Visit one and 18-month follow-up will include an integrated and cohesive battery of vascular and cognitive measurements, including peripheral endothelial function (flow-mediated dilation, laser speckle contrast imaging), retinal and cerebrovascular hemodynamics (dynamic vessel retinal analysis, functional near-infrared spectroscopy), and fluid and crystalized intelligence (NIH-Toolbox, n-back). Multiple logistic regression will be used for primary longitudinal data analysis to determine whether COVID-19 related impairment in neurovascular coupling and increases in white matter hyperintensity burden contribute to progression to dementia.
Collapse
Affiliation(s)
- Cameron D. Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Lauren Olay
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Ann L. Olson
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Guangpu Li
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Veronica Galvan
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
| | - Angelia C. Kirkpatrick
- Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Priya Balasubramanian
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Medicine, Cardiovascular Section, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Doctoral School of Basic and Translational Medicine/Departments of Public Health, International Training Program in Geroscience, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Calin I. Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, United States of America
- Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Neurosurgery, Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States of America
| |
Collapse
|
21
|
Wang D, Brady T, Santhanam L, Gerecht S. The extracellular matrix mechanics in the vasculature. NATURE CARDIOVASCULAR RESEARCH 2023; 2:718-732. [PMID: 39195965 DOI: 10.1038/s44161-023-00311-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Accepted: 06/20/2023] [Indexed: 08/29/2024]
Abstract
Mechanical stimuli from the extracellular matrix (ECM) modulate vascular differentiation, morphogenesis and dysfunction of the vasculature. With innovation in measurements, we can better characterize vascular microenvironment mechanics in health and disease. Recent advances in material sciences and stem cell biology enable us to accurately recapitulate the complex and dynamic ECM mechanical microenvironment for in vitro studies. These biomimetic approaches help us understand the signaling pathways in disease pathologies, identify therapeutic targets, build tissue replacement and activate tissue regeneration. This Review analyzes how ECM mechanics regulate vascular homeostasis and dysfunction. We highlight approaches to examine ECM mechanics at tissue and cellular levels, focusing on how mechanical interactions between cells and the ECM regulate vascular phenotype, especially under certain pathological conditions. Finally, we explore the development of biomaterials to emulate, measure and alter the physical microenvironment of pathological ECM to understand cell-ECM mechanical interactions toward the development of therapeutics.
Collapse
Affiliation(s)
- Dafu Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Travis Brady
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Lakshmi Santhanam
- Department of Anesthesiology and Critical Care Medicine and Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Sharon Gerecht
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
22
|
Zhor C, Wafaa L, Ghzaiel I, Kessas K, Zarrouk A, Ksila M, Ghrairi T, Latruffe N, Masmoudi-Kouki O, El Midaoui A, Vervandier-Fasseur D, Hammami M, Lizard G, Vejux A, Kharoubi O. Effects of polyphenols and their metabolites on age-related diseases. Biochem Pharmacol 2023:115674. [PMID: 37414102 DOI: 10.1016/j.bcp.2023.115674] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 06/26/2023] [Accepted: 06/27/2023] [Indexed: 07/08/2023]
Abstract
Aging contributes to the progressive loss of cellular biological functions and increases the risk of age-related diseases. Cardiovascular diseases, some neurological disorders and cancers are generally classified as age-related diseases that affect the lifespan of individuals. These diseases result from the accumulation of cellular damage and reduced activity of protective stress response pathways, which can lead to inflammation and oxidative stress, which play a key role in the aging process. There is now increasing interest in the therapeutic effects of edible plants for the prevention of various diseases, including those associated with aging. It has become clear that the beneficial effects of these foods are due, at least in part, to the high concentration of bioactive phenolic compounds with low side effects. Antioxidants are the most abundant, and their high consumption in the Mediterranean diet has been associated with slower ageing in humans. Extensive human dietary intervention studies strongly suggest that polyphenol supplementation protects against the development of degenerative diseases, especially in the elderly. In this review, we present data on the biological effects of plant polyphenols in the context of their relevance to human health, ageing and the prevention of age-related diseases.
Collapse
Affiliation(s)
- Chouari Zhor
- University Oran 1 ABB: laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences. Oran Algeria.
| | - Lounis Wafaa
- University Oran 1 ABB: laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences. Oran Algeria.
| | - Imen Ghzaiel
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, 21000 Dijon, France; University of Monastir: Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia; University Tunis-El Manar, Faculty of Sciences of Tunis, 2092 Tunis, Tunisia.
| | - Khadidja Kessas
- University Oran 1 ABB: laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences. Oran Algeria.
| | - Amira Zarrouk
- University of Monastir: Faculty of Medicine, LR12ES05, Lab-NAFS 'Nutrition - Functional Food & Vascular Health', 5000 Monastir, Tunisia; University of Sousse: Faculty of Medicine, Sousse, Tunisia.
| | - Mohamed Ksila
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, 21000 Dijon, France; University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMolecules, LR18ES03, Department of Biology, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Taoufik Ghrairi
- University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMolecules, LR18ES03, Department of Biology, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Norbert Latruffe
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, 21000 Dijon, France.
| | - Olfa Masmoudi-Kouki
- University Tunis-El Manar, Loboratory of Neurophysiology, Cellular Physiopathology and Valorisation of BioMolecules, LR18ES03, Department of Biology, Faculty of Sciences, 2092 Tunis, Tunisia.
| | - Adil El Midaoui
- Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Canada.
| | - Dominique Vervandier-Fasseur
- Institut de Chimie Moléculaire de l'Université de Bourgogne, ICMUB-UMR CNRS 6302, University Bourgogne Franche-Comté, 9, avenue A. Savary, 21078 Dijon Cedex, France.
| | - Mohamed Hammami
- Lab-NAFS 'Nutrition-Functional Food & Vascular Health', Faculty of Medicine, University of Monastir, LR12ES05, 5000 Monastir, Tunisia.
| | - Gérard Lizard
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, 21000 Dijon, France.
| | - Anne Vejux
- University Bourgogne Franche-Comté, Team 'Biochemistry of the Peroxisome, Inflammation and Lipid Metabolism' EA7270/Inserm, 21000 Dijon, France.
| | - Omar Kharoubi
- University Oran 1 ABB: laboratory of Experimental Biotoxicology, Biodepollution and Phytoremediation, Faculty of Life and Natural Sciences. Oran Algeria.
| |
Collapse
|
23
|
Owens CD, Pinto CB, Detwiler S, Mukli P, Peterfi A, Szarvas Z, Hoffmeister JR, Galindo J, Noori J, Kirkpatrick AC, Dasari TW, James J, Tarantini S, Csiszar A, Ungvari Z, Prodan CI, Yabluchanskiy A. Cerebral small vessel disease pathology in COVID-19 patients: A systematic review. Ageing Res Rev 2023; 88:101962. [PMID: 37224885 PMCID: PMC10202464 DOI: 10.1016/j.arr.2023.101962] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 05/20/2023] [Accepted: 05/21/2023] [Indexed: 05/26/2023]
Abstract
Cerebral small vessel disease (CSVD) is the leading cause of vascular cognitive impairment and is associated with COVID-19. However, contributing factors that often accompany CSVD pathology in COVID-19 patients may influence the incidence of cerebrovascular complications. Thus, a mechanism linking COVID-19 and CSVD has yet to be uncovered and differentiated from age-related comorbidities (i.e., hypertension), and medical interventions during acute infection. We aimed to evaluate CSVD in acute and recovered COVID-19 patients and to differentiate COVID-19-related cerebrovascular pathology from the above-mentioned contributing factors by assessing the localization of microbleeds and ischemic lesions/infarctions in the cerebrum, cerebellum, and brainstem. A systematic search was performed in December 2022 on PubMed, Web of Science, and Embase using a pre-established search criterion related to history of, or active COVID-19 with CSVD pathology in adults. From a pool of 161 studies, 59 met eligibility criteria and were included. Microbleeds and ischemic lesions had a strong predilection for the corpus callosum and subcortical/deep white matter in COVID-19 patients, suggesting a distinct CSVD pathology. These findings have important implications for clinical practice and biomedical research as COVID-19 may independently, and through exacerbation of age-related mechanisms, contribute to increased incidence of CSVD.
Collapse
Affiliation(s)
- Cameron D Owens
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Camila Bonin Pinto
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Sam Detwiler
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Peter Mukli
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Anna Peterfi
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zsofia Szarvas
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Jordan R Hoffmeister
- Department of Psychiatry and Behavioral Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Juliette Galindo
- Department of Psychiatry and Behavioral Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jila Noori
- Dean McGee Eye Institute, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Angelia C Kirkpatrick
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Tarun W Dasari
- Cardiovascular Section, Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Judith James
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA; Department of Internal Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Stefano Tarantini
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Anna Csiszar
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; International Training Program in Geroscience, Doctoral School of Basic and Translational Medicine/Departments of Public Health, Translational Medicine and Physiology, Semmelweis University, Budapest, Hungary; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Calin I Prodan
- Veterans Affairs Medical Center, Oklahoma City, OK, USA; Department of Neurology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Andriy Yabluchanskiy
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; The Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
| |
Collapse
|
24
|
Phua TJ. Understanding human aging and the fundamental cell signaling link in age-related diseases: the middle-aging hypovascularity hypoxia hypothesis. FRONTIERS IN AGING 2023; 4:1196648. [PMID: 37384143 PMCID: PMC10293850 DOI: 10.3389/fragi.2023.1196648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/23/2023] [Indexed: 06/30/2023]
Abstract
Aging-related hypoxia, oxidative stress, and inflammation pathophysiology are closely associated with human age-related carcinogenesis and chronic diseases. However, the connection between hypoxia and hormonal cell signaling pathways is unclear, but such human age-related comorbid diseases do coincide with the middle-aging period of declining sex hormonal signaling. This scoping review evaluates the relevant interdisciplinary evidence to assess the systems biology of function, regulation, and homeostasis in order to discern and decipher the etiology of the connection between hypoxia and hormonal signaling in human age-related comorbid diseases. The hypothesis charts the accumulating evidence to support the development of a hypoxic milieu and oxidative stress-inflammation pathophysiology in middle-aged individuals, as well as the induction of amyloidosis, autophagy, and epithelial-to-mesenchymal transition in aging-related degeneration. Taken together, this new approach and strategy can provide the clarity of concepts and patterns to determine the causes of declining vascularity hemodynamics (blood flow) and physiological oxygenation perfusion (oxygen bioavailability) in relation to oxygen homeostasis and vascularity that cause hypoxia (hypovascularity hypoxia). The middle-aging hypovascularity hypoxia hypothesis could provide the mechanistic interface connecting the endocrine, nitric oxide, and oxygen homeostasis signaling that is closely linked to the progressive conditions of degenerative hypertrophy, atrophy, fibrosis, and neoplasm. An in-depth understanding of these intrinsic biological processes of the developing middle-aged hypoxia could provide potential new strategies for time-dependent therapies in maintaining healthspan for healthy lifestyle aging, medical cost savings, and health system sustainability.
Collapse
Affiliation(s)
- Teow J. Phua
- Molecular Medicine, NSW Health Pathology, John Hunter Hospital, Newcastle, NSW, Australia
| |
Collapse
|
25
|
Berkowicz P, Totoń-Żurańska J, Kwiatkowski G, Jasztal A, Csípő T, Kus K, Tyrankiewicz U, Orzyłowska A, Wołkow P, Tóth A, Chlopicki S. Accelerated ageing and coronary microvascular dysfunction in chronic heart failure in Tgαq*44 mice. GeroScience 2023; 45:1619-1648. [PMID: 36692592 PMCID: PMC10400753 DOI: 10.1007/s11357-022-00716-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 12/12/2022] [Indexed: 01/25/2023] Open
Abstract
Age represents a major risk factor in heart failure (HF). However, the mechanisms linking ageing and HF are not clear. We aimed to identify the functional, morphological and transcriptomic changes that could be attributed to cardiac ageing in a model of slowly progressing HF in Tgαq*44 mice in reference to the cardiac ageing process in FVB mice. In FVB mice, ageing resulted in the impairment of diastolic cardiac function and in basal coronary flow (CF), perivascular and interstitial fibrosis without changes in the cardiac activity of angiotensin-converting enzyme (ACE) or aldosterone plasma concentration. In Tgαq*44 mice, HF progression was featured by the impairment of systolic and diastolic cardiac function and in basal CF that was associated with a distinct rearrangement of the capillary architecture, pronounced perivascular and interstitial fibrosis, progressive activation of cardiac ACE and systemic angiotensin-aldosterone-dependent pathways. Interestingly, cardiac ageing genes and processes were represented in Tgαq*44 mice not only in late but also in early phases of HF, as evidenced by cardiac transcriptome analysis. Thirty-four genes and 8 biological processes, identified as being ageing related, occurred early and persisted along HF progression in Tgαq*44 mice and were mostly associated with extracellular matrix remodelling and fibrosis compatible with perivascular fibrosis resulting in coronary microvascular dysfunction (CMD) in Tgαq*44 mice. In conclusion, accelerated and persistent cardiac ageing contributes to the pathophysiology of chronic HF in Tgαq*44 mice. In particular, prominent perivascular fibrosis of microcirculation resulting in CMD represents an accelerated cardiac ageing phenotype that requires targeted treatment in chronic HF.
Collapse
Affiliation(s)
- Piotr Berkowicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Justyna Totoń-Żurańska
- Centre for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Agnieszka Jasztal
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Tamás Csípő
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
- Department of Public Health, Faculty of Medicine, Semmelweis University, Budapest, Hungary
| | - Kamil Kus
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Urszula Tyrankiewicz
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland
| | - Anna Orzyłowska
- Department of Neurosurgery and Paediatric Neurosurgery, Medical University of Lublin, Lublin, Poland
| | - Paweł Wołkow
- Centre for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Attila Tóth
- Division of Clinical Physiology, Department of Cardiology, Faculty of Medicine, University of Debrecen, Debrecen, Hungary
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Krakow, Poland.
- Faculty of Medicine, Chair of Pharmacology, Jagiellonian University Medical College, Krakow, Poland.
| |
Collapse
|
26
|
Zhang X, An H, Chen Y, Shu N. Neurobiological Mechanisms of Cognitive Decline Correlated with Brain Aging. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1419:127-146. [PMID: 37418211 DOI: 10.1007/978-981-99-1627-6_10] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Cognitive decline has emerged as one of the greatest health threats of old age. Meanwhile, aging is the primary risk factor for Alzheimer's disease (AD) and other prevalent neurodegenerative disorders. Developing therapeutic interventions for such conditions demands a greater understanding of the processes underlying normal and pathological brain aging. Despite playing an important role in the pathogenesis and incidence of disease, brain aging has not been well understood at a molecular level. Recent advances in the biology of aging in model organisms, together with molecular- and systems-level studies of the brain, are beginning to shed light on these mechanisms and their potential roles in cognitive decline. This chapter seeks to integrate the knowledge about the neurological mechanisms of age-related cognitive changes that underlie aging.
Collapse
Affiliation(s)
- Xiaxia Zhang
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Haiting An
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
- Beijing Neurosurgical Institute, Beijing Tian Tan Hospital, Capital Medical University, Beijing, China
| | - Yuan Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China
- Beijing Aging Brain Rejuvenation Initiative (BABRI) Centre, Beijing Normal University, Beijing, China
| | - Ni Shu
- State Key Laboratory of Cognitive Neuroscience and Learning, Faculty of Psychology, Beijing Normal University, Beijing, China.
| |
Collapse
|
27
|
Clara FM, Alfie J, Scandurra A, Fita M. The Diastolic Oscillation Amplitude Used as an Arterial Aging Indicator. Vasc Health Risk Manag 2023; 19:193-200. [PMID: 37038502 PMCID: PMC10082575 DOI: 10.2147/vhrm.s381452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 03/27/2023] [Indexed: 04/12/2023] Open
Abstract
Introduction The compliance of the distal arteries depends on their vasoconstrictor tone and distensibility and is sensitive to endothelial function and aging. C2, a component of the Windkessel model, is a measure of distal arterial compliance, and establishes the magnitude of the pressure rise during early diastole. It is calculated from the diastolic portion of the radial pulse wave using sophisticated analyses. C2 is used as a cardiovascular risk indicator since it decreases with aging, high blood pressure, and diabetes. Here, we propose an alternative method to assess the distal arteries distensibility by measuring the amplitude of the oscillation that occurs at the beginning of diastole. Methods Peripheral pulse wave was evaluated noninvasively by applanation tonometry in 511 individuals (264 women) aged between 13 and 70 years. Diastolic amplitude (DA) was measured as the peak-to-peak amplitude of the diastolic oscillation. Radial augmentation index (RAIx) and pulse wave velocity (PWV) were also calculated. Results DA decreased approximately 2% per decade of life between 16 and 70 years from 19% to 7%, and was higher in men than in women (p<0.0001). Linear regression analysis identified RAIx as the strongest predictor of AD (p<0.0001), followed by age and height. Sex modified the age-related decrease in DA (p< 0.001). By applying the method to measure DA from previously published data, we found a strong linear correlation with C2. Conclusion DA decreased linearly with age in a reciprocal manner to the increase in radial augmentation index, was greater in men than women, and was independent of blood pressure and heart rate, as previously reported for C2. We propose that measuring DA could provide an alternative index to evaluate distal arterial compliance and aging.
Collapse
Affiliation(s)
- Fernando M Clara
- Instituto de Investigaciones Científicas y Tecnológicas en Electrónica, Universidad Nacional de Mar del Plata/ CONICET, Mar del Plata, Argentina
- Unidad Cardiometabólica, Centro de Especialidades Médicas Ambulatorias, Mar del Plata, Argentina
- Correspondence: Fernando M Clara, ICyTE, UNMDP/CONICET, Av. Juan B. Justo 4302, Mar del Plata, B7600, Argentina, Tel +54 0223 481-0046, Email
| | - José Alfie
- Unidad de Hipertensión Arterial, Servicio de Clínica Médica, Hospital Italiano de Buenos Aires, Ciudad de Buenos Aires, Argentina
| | - Adriana Scandurra
- Instituto de Investigaciones Científicas y Tecnológicas en Electrónica, Universidad Nacional de Mar del Plata/ CONICET, Mar del Plata, Argentina
| | - Mariana Fita
- Unidad Cardiometabólica, Centro de Especialidades Médicas Ambulatorias, Mar del Plata, Argentina
| |
Collapse
|
28
|
Age-Related Changes in Skeletal Muscle Oxygen Utilization. J Funct Morphol Kinesiol 2022; 7:jfmk7040087. [PMID: 36278748 PMCID: PMC9590092 DOI: 10.3390/jfmk7040087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 09/07/2022] [Accepted: 09/08/2022] [Indexed: 11/17/2022] Open
Abstract
The cardiovascular and skeletal muscle systems are intrinsically interconnected, sharing the goal of delivering oxygen to metabolically active tissue. Deficiencies within those systems that affect oxygen delivery to working tissues are a hallmark of advancing age. Oxygen delivery and utilization are reflected as muscle oxygen saturation (SmO2) and are assessed using near-infrared resonance spectroscopy (NIRS). SmO2 has been observed to be reduced by ~38% at rest, ~24% during submaximal exercise, and ~59% during maximal exercise with aging (>65 y). Furthermore, aging prolongs restoration of SmO2 back to baseline by >50% after intense exercise. Regulatory factors that contribute to reduced SmO2 with age include blood flow, capillarization, endothelial cells, nitric oxide, and mitochondrial function. These mechanisms are governed by reactive oxygen species (ROS) at the cellular level. However, mishandling of ROS with age ultimately leads to alterations in structure and function of the regulatory factors tasked with maintaining SmO2. The purpose of this review is to provide an update on the current state of the literature regarding age-related effects in SmO2. Furthermore, we attempt to bridge the gap between SmO2 and associated underlying mechanisms affected by aging.
Collapse
|
29
|
Caporarello N, Lee J, Pham TX, Jones DL, Guan J, Link PA, Meridew JA, Marden G, Yamashita T, Osborne CA, Bhagwate AV, Huang SK, Nicosia RF, Tschumperlin DJ, Trojanowska M, Ligresti G. Dysfunctional ERG signaling drives pulmonary vascular aging and persistent fibrosis. Nat Commun 2022; 13:4170. [PMID: 35879310 PMCID: PMC9314350 DOI: 10.1038/s41467-022-31890-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/04/2022] [Indexed: 01/18/2023] Open
Abstract
Vascular dysfunction is a hallmark of chronic diseases in elderly. The contribution of the vasculature to lung repair and fibrosis is not fully understood. Here, we performed an epigenetic and transcriptional analysis of lung endothelial cells (ECs) from young and aged mice during the resolution or progression of bleomycin-induced lung fibrosis. We identified the transcription factor ETS-related gene (ERG) as putative orchestrator of lung capillary homeostasis and repair, and whose function is dysregulated in aging. ERG dysregulation is associated with reduced chromatin accessibility and maladaptive transcriptional responses to injury. Loss of endothelial ERG enhances paracrine fibroblast activation in vitro, and impairs lung fibrosis resolution in young mice in vivo. scRNA-seq of ERG deficient mouse lungs reveales transcriptional and fibrogenic abnormalities resembling those associated with aging and human lung fibrosis, including reduced number of general capillary (gCap) ECs. Our findings demonstrate that lung endothelial chromatin remodeling deteriorates with aging leading to abnormal transcription, vascular dysrepair, and persistent fibrosis following injury.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jisu Lee
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Tho X Pham
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Dakota L Jones
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiazhen Guan
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Patrick A Link
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey A Meridew
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Grace Marden
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Takashi Yamashita
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Collin A Osborne
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Aditya V Bhagwate
- Department of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN, USA
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Roberto F Nicosia
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | | | - Maria Trojanowska
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
30
|
Fisher RA, Miners JS, Love S. Pathological changes within the cerebral vasculature in Alzheimer's disease: New perspectives. Brain Pathol 2022; 32:e13061. [PMID: 35289012 PMCID: PMC9616094 DOI: 10.1111/bpa.13061] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 02/11/2022] [Accepted: 02/21/2022] [Indexed: 12/14/2022] Open
Abstract
Cerebrovascular disease underpins vascular dementia (VaD), but structural and functional changes to the cerebral vasculature contribute to disease pathology and cognitive decline in Alzheimer's disease (AD). In this review, we discuss the contribution of cerebral amyloid angiopathy and non‐amyloid small vessel disease in AD, and the accompanying changes to the density, maintenance and remodelling of vessels (including alterations to the composition and function of the cerebrovascular basement membrane). We consider how abnormalities of the constituent cells of the neurovascular unit – particularly of endothelial cells and pericytes – and impairment of the blood‐brain barrier (BBB) impact on the pathogenesis of AD. We also discuss how changes to the cerebral vasculature are likely to impair Aβ clearance – both intra‐periarteriolar drainage (IPAD) and transport of Aβ peptides across the BBB, and how impaired neurovascular coupling and reduced blood flow in relation to metabolic demand increase amyloidogenic processing of APP and the production of Aβ. We review the vasoactive properties of Aβ peptides themselves, and the probable bi‐directional relationship between vascular dysfunction and Aβ accumulation in AD. Lastly, we discuss recent methodological advances in transcriptomics and imaging that have provided novel insights into vascular changes in AD, and recent advances in assessment of the retina that allow in vivo detection of vascular changes in the early stages of AD.
Collapse
Affiliation(s)
- Robert A Fisher
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - J Scott Miners
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| | - Seth Love
- Dementia Research Group, University of Bristol Medical School, Bristol, UK
| |
Collapse
|
31
|
Deciphering the endometrial niche of human thin endometrium at single-cell resolution. Proc Natl Acad Sci U S A 2022; 119:2115912119. [PMID: 35169075 PMCID: PMC8872762 DOI: 10.1073/pnas.2115912119] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2022] [Indexed: 12/16/2022] Open
Abstract
Thin endometrium is the most common reason for uterine infertility and refractory gynecological diseases due to its complexity in pathogenesis and adverse pregnancy outcomes. Here, we profile cells from normal and thin endometrium at single-cell resolution to investigate the sophisticated alterations in the local microenvironment that occur in thin endometrium. Increased cellular senescence, collagen overdeposition, and significant down-regulation of gene expression related to cell proliferation are observed and confirmed. Moreover, we demonstrate aberrant activation of the SEMA3 pathway accompanied by dampened EGF, PTN, and TWEAK signaling pathways in thin endometrium. These findings aid in understanding the mechanisms of thin endometrium and provide new tools to rejuvenate the atrophic endometrium for female fertility preservation and successful pregnancy. Thin endometrium has been widely recognized as a critical cause of infertility, recurrent pregnancy loss, and placental abnormalities; however, access to effective treatment is a formidable challenge due to the rudimentary understanding of the pathogenesis of thin endometrium. Here, we profiled the transcriptomes of human endometrial cells at single-cell resolution to characterize cell types, their communications, and the underlying mechanism of endometrial growth in normal and thin endometrium during the proliferative phase. Stromal cells were the most abundant cell type in the endometrium, with a subpopulation of proliferating stromal cells whose cell cycle signaling pathways were compromised in thin endometrium. Both single-cell RNA sequencing and experimental verification revealed cellular senescence in the stroma and epithelium accompanied by collagen overdeposition around blood vessels. Moreover, decreased numbers of macrophages and natural killer cells further exacerbated endometrial thinness. In addition, our results uncovered aberrant SEMA3, EGF, PTN, and TWEAK signaling pathways as causes for the insufficient proliferation of the endometrium. Together, these data provide insight into therapeutic strategies for endometrial regeneration and growth to treat thin endometrium.
Collapse
|
32
|
Hachmo Y, Hadanny A, Mendelovic S, Hillman P, Shapira E, Landau G, Gattegno H, Zrachya A, Daniel-Kotovsky M, Catalogna M, Fishlev G, Lang E, Polak N, Doenyas K, Friedman M, Zemel Y, Bechor Y, Efrati S. The effect of hyperbaric oxygen therapy on the pathophysiology of skin aging: a prospective clinical trial. Aging (Albany NY) 2021; 13:24500-24510. [PMID: 34784294 PMCID: PMC8660605 DOI: 10.18632/aging.203701] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/20/2021] [Indexed: 11/25/2022]
Abstract
INTRODUCTION Skin biopsies can be used to evaluate physiological effects of aging targeted intervention at the tissue/cellular levels. Recent clinical trials have shown that hyperbaric oxygen therapy (HBOT) can target aging hallmarks, including telomere shortening, senescent cells clearance and angiogenesis. The aim of this study was to evaluate the effects of HBOT on the skin of a normal, non-pathological, aging population. METHODS The study was performed as a prospective clinical trial. After signing informed consent and undergoing baseline evaluations, the subjects were assigned to a three-month control period followed by three months of HBOT daily sessions. Skin biopsies were taken at baseline, after three months of no intervention (control) and 1-2 weeks following the last HBOT session. Trichrome, Orecin, lipofuscin and CD31 staining were used to evaluate collagen fibers, elastic fibers, senescent cells and blood vessels, respectively. RESULTS Out of the cohort of 70 participants in the normal aging population study, thirteen male patients (age 68.07±2.5y) gave consent for repeated skin biopsies. Following HBOT, there was a significant increase in collagen density (p<0.001, effect size(es)=1.10), elastic fiber length (p<0.0001, es=2.71) and the number of blood vessels (p=0.02, es=1.00). There was a significant decrease in fiber fragmentation (p=0.012) and in tissue senescent cells (p=0.03, es=0.84) post-HBOT. No changes were noted in elastic fiber density or thickness. CONCLUSIONS The study indicates, for the first time in humans, that HBOT can significantly modulate the pathophysiology of the skin aging in a healthy aging population. The demonstrated mechanisms include angiogenesis and senescent cell clearance.
Collapse
Affiliation(s)
- Yafit Hachmo
- Research and Development Unit, Shamir Medical Center, Zerifin, Israel
| | - Amir Hadanny
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Bar Ilan University, Ramat-Gan, Israel
| | - Sonia Mendelovic
- Pathology Department, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Pnina Hillman
- Pathology Department, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Eyal Shapira
- Plastic Surgery Department, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Geva Landau
- Plastic Surgery Department, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Hadar Gattegno
- Research and Development Unit, Shamir Medical Center, Zerifin, Israel
| | - Avi Zrachya
- Research and Development Unit, Shamir Medical Center, Zerifin, Israel
| | - Malka Daniel-Kotovsky
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Merav Catalogna
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Gregory Fishlev
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Erez Lang
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Nir Polak
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Keren Doenyas
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Mony Friedman
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Yonatan Zemel
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Yair Bechor
- The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel
| | - Shai Efrati
- Research and Development Unit, Shamir Medical Center, Zerifin, Israel.,The Sagol Center for Hyperbaric Medicine and Research, Shamir (Assaf-Harofeh) Medical Center, Zerifin, Israel.,Sackler School of Medicine, Tel-Aviv University, Tel-Aviv, Israel.,Sagol School of Neuroscience, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
33
|
Faria D, Lee J, van der Hoef T, Mejía-Rentería H, Echavarria-Pinto M, Baptista S, Cerrato E, Garcia-Garcia H, Davies J, Onuma Y, Samady H, Piek JJ, Serruys PW, Lerman A, Escaned J. Age and functional relevance of coronary stenosis: a post hoc analysis of the ADVISE II trial. EUROINTERVENTION 2021; 17:757-764. [PMID: 33720019 PMCID: PMC9724935 DOI: 10.4244/eij-d-20-01163] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
BACKGROUND The influence of age-dependent changes on fractional flow reserve (FFR) or instantaneous wave-free ratio (iFR) and the response to pharmacological hyperaemia has not been investigated. AIMS We investigated the impact of age on these indices. METHODS This is a post hoc analysis of the ADVISE II trial, including a total of 690 pressure recordings (in 591 patients). Age-dependent correlations with FFR and iFR were calculated and adjusted for stenosis severity. Patients were stratified into three age terciles. The hyperaemic response to adenosine, calculated as the difference between resting and hyperaemic pressure ratios, and the prevalence of FFR-iFR discordance were assessed. RESULTS Age correlated positively with FFR (r=0.08, 95% CI: 0.01 to 0.15, p=0.015), but not with iFR (r=-0.03, 95% CI: -0.11 to 0.04, p=0.411). The hyperaemic response to adenosine decreased with patient age (0.12±0.07, 0.11±0.06, 0.09±0.05, for the 1st [33-58 years], 2nd [59-69 years] and 3rd [70-94 years] age tertiles, respectively, p<0.001) and showed significant correlation with age (r=-0.14, 95% CI: -0.21 to -0.06, p<0.001). The proportion of patients with FFR ≤0.80+iFR >0.89 discordance doubled in the first age tercile (14.1% vs 7.1% vs 7.0%, p=0.005). CONCLUSIONS The hyperaemic response of the microcirculation to adenosine administration is age dependent. FFR values increase with patient age, while iFR values remain constant across the age spectrum. These findings contribute to explaining differences observed in functional stenosis classification with hyperaemic and non-hyperaemic coronary indices.
Collapse
Affiliation(s)
- Daniel Faria
- Interventional Cardiology Unit, Hospital Clinico San Carlos IDISSC, Complutense University of Madrid, Madrid, Spain,Department of Cardiology, Hospital Professor Doutor Fernando Fonseca, Amadora, Portugal
| | - Joo Lee
- Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Tim van der Hoef
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Interventional Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Hernán Mejía-Rentería
- Interventional Cardiology Unit, Hospital Clinico San Carlos IDISSC, Complutense University of Madrid, Madrid, Spain
| | - Mauro Echavarria-Pinto
- Interventional Cardiology Unit, Hospital Clinico San Carlos IDISSC, Complutense University of Madrid, Madrid, Spain,Hospital General ISSSTE Querétaro, Facultad de Medicina, Universidad Autónoma de Querétaro, Santiago de Querétaro, México
| | - Sérgio Baptista
- Department of Cardiology, Hospital Professor Doutor Fernando Fonseca, Amadora, Portugal,University Clinic of Cardiology, Faculty of Medicine at University of Lisbon, Lisbon, Portugal
| | - Enrico Cerrato
- Interventional Cardiology Unit, San Luigi Gonzaga University Hospital, Orbassano, Italy,Rivoli Infermi Hospital, Turin, Italy
| | - Hector Garcia-Garcia
- Interventional Cardiology, Medstar Washington Hospital Center, Washington, DC, USA
| | - Justin Davies
- Faculty of Medicine, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Yoshinobu Onuma
- Department of Interventional Cardiology, Thoraxcenter, Erasmus Medical Center, Rotterdam, the Netherlands
| | - Habib Samady
- Department of Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Jan J. Piek
- Amsterdam UMC, University of Amsterdam, Heart Center, Department of Interventional Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam, the Netherlands
| | - Patrick W. Serruys
- Department of Cardiology, National University of Ireland, Galway, Ireland
| | - Amir Lerman
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN, USA
| | - Javier Escaned
- Interventional Cardiology, Hospital Clinico San Carlos, Calle del Prof Martín Lagos, 28040 Madrid, Spain
| |
Collapse
|
34
|
Herrod PJJ, Atherton PJ, Smith K, Williams JP, Lund JN, Phillips BE. Six weeks of high-intensity interval training enhances contractile activity induced vascular reactivity and skeletal muscle perfusion in older adults. GeroScience 2021; 43:2667-2678. [PMID: 34562202 PMCID: PMC8602610 DOI: 10.1007/s11357-021-00463-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 09/18/2021] [Indexed: 11/28/2022] Open
Abstract
Impairments in muscle microvascular function are associated with the pathogenesis of sarcopenia and cardiovascular disease. High-intensity interval training (HIIT) is an intervention by which a myriad of beneficial skeletal muscle/cardiovascular adaptations have been reported across age, including capillarisation and improved endothelial function. Herein, we hypothesised that HIIT would enhance muscle microvascular blood flow and vascular reactivity to acute contractile activity in older adults, reflecting HIIT-induced vascular remodelling. In a randomised controlled-trial, twenty-five healthy older adults aged 65–85 years (mean BMI 27.0) were randomised to 6-week HIIT or a no-intervention control period of an equal duration. Measures of microvascular responses to a single bout of muscle contractions (i.e. knee extensions) were made in the m. vastus lateralis using contrast-enhanced ultrasound during a continuous intravenous infusion of Sonovue™ contrast agent, before and after the intervention period, with concomitant assessments of cardiorespiratory fitness and resting blood pressure. HIIT led to improvements in anaerobic threshold (13.2 ± 3.4 vs. 15.3 ± 3.8 ml/kg/min, P < 0.001), dynamic exercise capacity (145 ± 60 vs. 159 ± 59 W, P < 0.001) and resting (systolic) blood pressure (142 ± 15 vs. 133 ± 11 mmHg, P < 0.01). Notably, HIIT elicited significant increases in microvascular blood flow responses to acute contractile activity (1.8 ± 0.63 vs. 2.3 ± 0.8 (arbitrary contrast units (AU), P < 0.01)), with no change in any of these parameters observed in the control group. Six weeks HIIT improves skeletal muscle microvascular responsiveness to acute contractile activity in the form of active hyperaemia-induced by a single bout of resistance exercise. These findings likely reflect reports of enhanced large vessel distensibility, improved endothelial function, and muscle capillarisation following HIIT. Moreover, our findings illustrate that HIIT may be effective in mitigating deleterious alterations in muscle microvascular mediated aspects of sarcopenia.
Collapse
Affiliation(s)
- Philip J J Herrod
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Royal Derby Hospital Centre, DE22 3DT, Derby, UK.,NIHR Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, UK.,Department of Anaesthetics and Surgery, Royal Derby Hospital, Derby, UK
| | - Philip J Atherton
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Royal Derby Hospital Centre, DE22 3DT, Derby, UK.,NIHR Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, UK
| | - Kenneth Smith
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Royal Derby Hospital Centre, DE22 3DT, Derby, UK.,NIHR Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, UK
| | - John P Williams
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Royal Derby Hospital Centre, DE22 3DT, Derby, UK.,NIHR Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, UK.,Department of Anaesthetics and Surgery, Royal Derby Hospital, Derby, UK
| | - Jonathan N Lund
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Royal Derby Hospital Centre, DE22 3DT, Derby, UK.,NIHR Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, UK.,Department of Anaesthetics and Surgery, Royal Derby Hospital, Derby, UK
| | - Bethan E Phillips
- Medical Research Council-Versus Arthritis Centre for Musculoskeletal Ageing Research, University of Nottingham, Royal Derby Hospital Centre, DE22 3DT, Derby, UK. .,NIHR Nottingham Biomedical Research Centre, Queens Medical Centre, Nottingham, UK.
| |
Collapse
|
35
|
Guéré C, Bigouret A, Nkengne A, Vié K, Gélis A, Dulong J, Lamartine J, Fromy B. In elderly Caucasian women, younger facial perceived age correlates with better forearm skin microcirculation reactivity. Skin Res Technol 2021; 27:1152-1161. [PMID: 34224600 DOI: 10.1111/srt.13080] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/24/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Visual and molecular changes occurring upon aging are rather well characterized. Still, aging signs show great significant inter-individual variations, and little is known concerning the link between perceived age and cutaneous microcirculation. MATERIALS AND METHODS To investigate this point, we recruited Caucasian women in their mid-50's to mid-70's and subsampled women looking older or younger than their age. We studied their facial skin color, as well as their microvascular reactivity to local heating assessed in the forearm skin. We also used skin biopsies from some of these women for gene expression or immunohistochemical analysis. RESULTS Clinical and instrumental analysis of skin color revealed that subjects who look 5 years younger differ only by a higher glowing complexion. Our most striking result is that subjects looking 5 years younger than their age present a higher microcirculation reactivity in forearm skin. Transcriptome comparison of skin samples from women looking older or younger than their age revealed 123 annotated transcripts differentially expressed, among which MYL9 relates to microcirculation. MYL9 is downregulated in the group of women looking younger than their real age. Microscopy shows that the labeling of MYL9 and CD31 are altered and heterogeneous with age, as is the morphology of microvessels. CONCLUSION Therefore, assessing generalized vascular reactivity in non-photo-exposed skin to focus on the intrinsic aging allows subtle discrimination of perceived age within elderly healthy subjects.
Collapse
Affiliation(s)
| | | | | | | | - Anthony Gélis
- Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique, UMR5305 CNRS - Université Claude Bernard, Lyon Cedex 07, France
| | - Joshua Dulong
- Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique, UMR5305 CNRS - Université Claude Bernard, Lyon Cedex 07, France
| | - Jérôme Lamartine
- Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique, UMR5305 CNRS - Université Claude Bernard, Lyon Cedex 07, France
| | - Bérengère Fromy
- Laboratoire de Biologie Tissulaire et d'ingénierie Thérapeutique, UMR5305 CNRS - Université Claude Bernard, Lyon Cedex 07, France
| |
Collapse
|
36
|
Zimmerman B, Rypma B, Gratton G, Fabiani M. Age-related changes in cerebrovascular health and their effects on neural function and cognition: A comprehensive review. Psychophysiology 2021; 58:e13796. [PMID: 33728712 PMCID: PMC8244108 DOI: 10.1111/psyp.13796] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/11/2021] [Accepted: 02/08/2021] [Indexed: 12/11/2022]
Abstract
The process of aging includes changes in cellular biology that affect local interactions between cells and their environments and eventually propagate to systemic levels. In the brain, where neurons critically depend on an efficient and dynamic supply of oxygen and glucose, age-related changes in the complex interaction between the brain parenchyma and the cerebrovasculature have effects on health and functioning that negatively impact cognition and play a role in pathology. Thus, cerebrovascular health is considered one of the main mechanisms by which a healthy lifestyle, such as habitual cardiorespiratory exercise and a healthful diet, could lead to improved cognitive outcomes with aging. This review aims at detailing how the physiology of the cerebral vascular system changes with age and how these changes lead to differential trajectories of cognitive maintenance or decline. This provides a framework for generating specific mechanistic hypotheses about the efficacy of proposed interventions and lifestyle covariates that contribute to enhanced cognitive well-being. Finally, we discuss the methodological implications of age-related changes in the cerebral vasculature for human cognitive neuroscience research and propose directions for future experiments aimed at investigating age-related changes in the relationship between physiology and cognitive mechanisms.
Collapse
Affiliation(s)
- Benjamin Zimmerman
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Bart Rypma
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, USA
- Department of Psychiatry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gabriele Gratton
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Monica Fabiani
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
- Department of Psychology, University of Illinois at Urbana-Champaign, Champaign, IL, USA
- Neuroscience Program, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| |
Collapse
|
37
|
Islam Y, Leach AG, Smith J, Pluchino S, Coxon CR, Sivakumaran M, Downing J, Fatokun AA, Teixidò M, Ehtezazi T. Physiological and Pathological Factors Affecting Drug Delivery to the Brain by Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2002085. [PMID: 34105297 PMCID: PMC8188209 DOI: 10.1002/advs.202002085] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 01/06/2021] [Indexed: 05/04/2023]
Abstract
The prevalence of neurological/neurodegenerative diseases, such as Alzheimer's disease is known to be increasing due to an aging population and is anticipated to further grow in the decades ahead. The treatment of brain diseases is challenging partly due to the inaccessibility of therapeutic agents to the brain. An increasingly important observation is that the physiology of the brain alters during many brain diseases, and aging adds even more to the complexity of the disease. There is a notion that the permeability of the blood-brain barrier (BBB) increases with aging or disease, however, the body has a defense mechanism that still retains the separation of the brain from harmful chemicals in the blood. This makes drug delivery to the diseased brain, even more challenging and complex task. Here, the physiological changes to the diseased brain and aged brain are covered in the context of drug delivery to the brain using nanoparticles. Also, recent and novel approaches are discussed for the delivery of therapeutic agents to the diseased brain using nanoparticle based or magnetic resonance imaging guided systems. Furthermore, the complement activation, toxicity, and immunogenicity of brain targeting nanoparticles as well as novel in vitro BBB models are discussed.
Collapse
Affiliation(s)
- Yamir Islam
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Andrew G. Leach
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- Division of Pharmacy and OptometryThe University of ManchesterStopford Building, Oxford RoadManchesterM13 9PTUK
| | - Jayden Smith
- Cambridge Innovation Technologies Consulting (CITC) LimitedSt. John's Innovation CentreCowley RoadCambridgeCB4 0WSUK
| | - Stefano Pluchino
- Department of Clinical NeurosciencesClifford Allbutt Building – Cambridge Biosciences Campus and NIHR Biomedical Research CentreUniversity of CambridgeHills RoadCambridgeCB2 0HAUK
| | - Christopher R. Coxon
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
- School of Engineering and Physical SciencesHeriot‐Watt UniversityWilliam Perkin BuildingEdinburghEH14 4ASUK
| | - Muttuswamy Sivakumaran
- Department of HaematologyPeterborough City HospitalEdith Cavell CampusBretton Gate PeterboroughPeterboroughPE3 9GZUK
| | - James Downing
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Amos A. Fatokun
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| | - Meritxell Teixidò
- Institute for Research in Biomedicine (IRB Barcelona)Barcelona Institute of Science and Technology (BIST)Baldiri Reixac 10Barcelona08028Spain
| | - Touraj Ehtezazi
- School of Pharmacy and Biomolecular SciencesLiverpool John Moores UniversityByrom StreetLiverpoolL3 3AFUK
| |
Collapse
|
38
|
Robbins CB, Grewal DS, Thompson AC, Yoon SP, Plassman BL, Fekrat S. Repeatability of Peripapillary Optical Coherence Tomography Angiography Parameters in Older Adults. JOURNAL OF VITREORETINAL DISEASES 2021; 5:239-246. [PMID: 37006509 PMCID: PMC9979033 DOI: 10.1177/2474126420953968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Purpose: This work assesses the intrasession repeatability of capillary perfusion density (CPD) and capillary flux index (CFI) measurements on peripapillary optical coherence tomography angiography (OCTA) in healthy eyes of older adults. Methods: In this cross-sectional study, healthy volunteers aged 50 years or older underwent 4.5 × 4.5 mm OCTA imaging centered on the optic nerve head using Zeiss Cirrus HD-5000 AngioPlex (Carl Zeiss Meditec). Two consecutive images were acquired in the same eye during a single study session. CPD and CFI were assessed using AngioPlex Software (version 11.0.0.29946) for the radial peripapillary capillary plexus (average over whole scan area) and 4 quadrants (superior, inferior, temporal, and nasal). CPD and CFI repeatability was assessed by intraclass correlation (ICC), mean interocular differences using 2-tailed t test, and association with age using generalized estimating equations. Results: A total of 150 images were acquired from 75 eyes of 47 patients. For CPD, ICC results ranged from 0.7160 (nasal CPD) to 0.9218 (average CPD). For CFI, ICC results ranged from 0.6167 (temporal CFI) to 0.8976 (inferior CFI). Temporal CFI was significantly different between right and left eyes of the same patient ( P = .03). CPD and CFI decreased with age in all analyses (average CPD β coefficient –0.00172, P < .001; average CFI β coefficient −0.00278, P < .001). Conclusions: Moderate to good repeatability was observed for most peripapillary OCTA metrics; temporal measurements were least repeatable for CPD and CFI. Peripapillary CPD and CFI decrease with age even beyond the fifth decade in healthy older adults.
Collapse
Affiliation(s)
- Cason B. Robbins
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Dilraj S. Grewal
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Atalie C. Thompson
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Stephen P. Yoon
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| | - Brenda L. Plassman
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Sharon Fekrat
- Department of Ophthalmology, Duke University School of Medicine, Durham, NC, USA
| |
Collapse
|
39
|
Abstract
The blood-brain barrier (BBB) protects the central nervous system (CNS) from unregulated exposure to the blood and its contents. The BBB also controls the blood-to-brain and brain-to-blood permeation of many substances, resulting in nourishment of the CNS, its homeostatic regulation and communication between the CNS and peripheral tissues. The cells forming the BBB communicate with cells of the brain and in the periphery. This highly regulated interface changes with healthy aging. Here, we review those changes, starting with morphology and disruption. Transporter changes include those for amyloid beta peptide, glucose and drugs. Brain fluid dynamics, pericyte health and basement membrane and glycocalyx compositions are all altered with healthy aging. Carrying the ApoE4 allele leads to an acceleration of most of the BBB's age-related changes. We discuss how alterations in the BBB that occur with healthy aging reflect adaptation to the postreproductive phase of life and may affect vulnerability to age-associated diseases.
Collapse
|
40
|
Ahmed KA, Kim K, Ricart K, Van Der Pol W, Qi X, Bamman MM, Behrens C, Fisher G, Boulton ME, Morrow C, O'Neal PV, Patel RP. Potential role for age as a modulator of oral nitrate reductase activity. Nitric Oxide 2021; 108:1-7. [PMID: 33321206 PMCID: PMC8085911 DOI: 10.1016/j.niox.2020.12.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 12/07/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023]
Abstract
Reduction of salivary nitrate to nitrite by oral nitrate reductase (NR) expressing bacteria has emerged as an integral pathway in regulating nitric oxide (NO) homeostasis and signaling. The oral microbiome is critical for this pathway. Variations in this pathway may underlie variable responses in the magnitude by which dietary or therapeutic nitrate modulates NO-signaling. The relationships between oral microbes and NR activity, and the factors that affect this relationship remain unclear however. Using a cross-sectional study design, the objective of this study was to determine the relationships between oral microbes and oral NR activity using a protocol that directly measures initial NR activity. Tongue swabs were collected from 28 subjects ranging in age from 21 to 73y. Initial NR activity showed a bell-shaped dependence with age, with activity peaking at ~40-50y and being lower but similar between younger (20-30y) and older (51-73) individuals. Microbiome relative abundance and diversity analyses, using 16s sequencing, demonstrated differences across age and identified both NR expressing and non-expressing bacteria in modulating initial NR activity. Finally, initial NR activity was measured in 3mo and 13mo old C57BL/6J mice. No differences in bacterial number were observed. However initial NR activity was significantly (80%) lower in 13mo old mice. Collectively, these data suggest that age is a variable in NR activity and may modulate responsiveness to dietary nitrate.
Collapse
Affiliation(s)
- Khandaker Ahtesham Ahmed
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Kiyoung Kim
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA; Department of Pharmacology & Experimental Therapeutics and the Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, MA, USA
| | - Karina Ricart
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - William Van Der Pol
- Center for Clinical and Translational Science, University of Alabama at Birmingham, Birmingham, USA
| | - Xiaoping Qi
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Marcas M Bamman
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christian Behrens
- Department of Nutrition Sciences, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gordon Fisher
- Department of Human Studies, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael E Boulton
- Department of Ophthalmology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Casey Morrow
- Department of Cell, Developmental and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Pamela V O'Neal
- College of Nursing, University of Alabama in Huntsville, Huntsville, AL, USA
| | - Rakesh P Patel
- Department of Pathology and Center for Free Radical Biology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
41
|
Hendricks NE. The microcirculation. SOUTHERN AFRICAN JOURNAL OF ANAESTHESIA AND ANALGESIA 2020. [DOI: 10.36303/sajaa.2020.26.6.s3.2540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
The microcirculation is responsible for the transfer of oxygen from the red blood cells in the capillaries to the cells to meet cellular energy requirements, support functional activity and remove carbon dioxide and waste. The microcirculation also assists in the regulation of vascular tone, solute exchange, the production of hormones, the inflammatory response and haemostasis. The identification of the endothelial glycocalyx and the assessment of the effects of disease, drugs and fluids on the microcirculation is the subject of ongoing research.
Collapse
|
42
|
Damodarasamy M, Vernon RB, Pathan JL, Keene CD, Day AJ, Banks WA, Reed MJ. The microvascular extracellular matrix in brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA). Fluids Barriers CNS 2020; 17:60. [PMID: 32993718 PMCID: PMC7525948 DOI: 10.1186/s12987-020-00219-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 09/09/2020] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND The microvasculature (MV) of brains with Alzheimer's disease neuropathologic change (ADNC) and cerebral amyloid angiopathy (CAA), in the absence of concurrent pathologies (e.g., infarctions, Lewy bodies), is incompletely understood. OBJECTIVE To analyze microvascular density, diameter and extracellular matrix (ECM) content in association with ADNC and CAA. METHODS We examined samples of cerebral cortex and isolated brain microvasculature (MV) from subjects with the National Institute on Aging-Alzheimer's Association (NIA-AA) designations of not-, intermediate-, or high ADNC and from subjects with no CAA and moderate-severe CAA. Cases for all groups were selected with no major (territorial) strokes, ≤ 1 microinfarct in screening sections, and no Lewy body pathology. MV density and diameter were measured from cortical brain sections. Levels of basement membrane (BM) ECM components, the protein product of TNF-stimulated gene-6 (TSG-6), and the ubiquitous glycosaminoglycan hyaluronan (HA) were assayed by western blots or HA ELISA of MV lysates. RESULTS We found no significant changes in MV density or diameter among any of the groups. Levels of BM laminin and collagen IV (col IV) were lower in MV isolated from the high ADNC vs. not-ADNC groups. In contrast, BM laminin was significantly higher in MV from the moderate-severe CAA vs. the no CAA groups. TSG-6 and HA content were higher in the presence of both high ADNC and CAA, whereas levels of BM fibronectin and perlecan were similar among all groups. CONCLUSIONS Cortical MV density and diameter are not appreciably altered by ADNC or CAA. TSG-6 and HA are increased in both ADNC and CAA, with laminin and col IV decreased in the BM of high ADNC, but laminin increased in moderate-severe CAA. These results show that changes in the ECM occur in AD and CAA, but independently of one another, and likely reflect on the regional functioning of the brain microvasculature.
Collapse
Affiliation(s)
- Mamatha Damodarasamy
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Robert B Vernon
- Matrix Biology Program, Benaroya Research Institute at Virginia Mason, Seattle, WA, USA
| | - Jasmine L Pathan
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
| | - C Dirk Keene
- Division of Neuropathology, Department of Pathology, University of Washington, Seattle, WA, USA
| | - Anthony J Day
- Wellcome Trust Centre for Cell-Matrix Research and Lydia Becker Institute of Immunology and Inflammation, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Sciences Centre, Manchester, UK
| | - William A Banks
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA
| | - May J Reed
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington, Seattle, WA, USA.
- Geriatric Research Education and Clinical Center, VA Puget Sound Health Care System, Seattle, WA, USA.
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington Harborview Medical Center, Seattle, WA, 98104, USA.
| |
Collapse
|
43
|
Yoon SK, Kim HN, Song SW. Associations of skeletal muscle mass with atherosclerosis and inflammatory markers in Korean adults. Arch Gerontol Geriatr 2020; 90:104163. [DOI: 10.1016/j.archger.2020.104163] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 05/29/2020] [Accepted: 06/09/2020] [Indexed: 12/25/2022]
|
44
|
Caporarello N, Meridew JA, Aravamudhan A, Jones DL, Austin SA, Pham TX, Haak AJ, Moo Choi K, Tan Q, Haresi A, Huang SK, Katusic ZS, Tschumperlin DJ, Ligresti G. Vascular dysfunction in aged mice contributes to persistent lung fibrosis. Aging Cell 2020; 19:e13196. [PMID: 32691484 PMCID: PMC7431829 DOI: 10.1111/acel.13196] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/28/2020] [Accepted: 06/21/2020] [Indexed: 12/23/2022] Open
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive disease thought to result from impaired lung repair following injury and is strongly associated with aging. While vascular alterations have been associated with IPF previously, the contribution of lung vasculature during injury resolution and fibrosis is not well understood. To compare the role of endothelial cells (ECs) in resolving and non-resolving models of lung fibrosis, we applied bleomycin intratracheally to young and aged mice. We found that injury in aged mice elicited capillary rarefaction, while injury in young mice resulted in increased capillary density. ECs from the lungs of injured aged mice relative to young mice demonstrated elevated pro-fibrotic and reduced vascular homeostasis gene expression. Among the latter, Nos3 (encoding the enzyme endothelial nitric oxide synthase, eNOS) was transiently upregulated in lung ECs from young but not aged mice following injury. Young mice deficient in eNOS recapitulated the non-resolving lung fibrosis observed in aged animals following injury, suggesting that eNOS directly participates in lung fibrosis resolution. Activation of the NO receptor soluble guanylate cyclase in human lung fibroblasts reduced TGFβ-induced pro-fibrotic gene and protein expression. Additionally, loss of eNOS in human lung ECs reduced the suppression of TGFβ-induced lung fibroblast activation in 2D and 3D co-cultures. Altogether, our results demonstrate that persistent lung fibrosis in aged mice is accompanied by capillary rarefaction, loss of EC identity, and impaired eNOS expression. Targeting vascular function may thus be critical to promote lung repair and fibrosis resolution in aging and IPF.
Collapse
Affiliation(s)
- Nunzia Caporarello
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Jeffrey A Meridew
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Aja Aravamudhan
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Dakota L Jones
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Susan A Austin
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | - Tho X Pham
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Andrew J Haak
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Kyoung Moo Choi
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Qi Tan
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Adil Haresi
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
| | - Steven K Huang
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zvonimir S Katusic
- Department of Anesthesiology and Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN, USA
| | | | - Giovanni Ligresti
- Department of Medicine, Boston University School of Medicine, Boston, MA, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
45
|
Dzeletovic B, Stratimirovic DJ, Stojic D, Djukic LJ. Linear and nonlinear analysis of dental pulp blood flow oscillations in ageing. Int Endod J 2020; 53:1033-1039. [PMID: 32281658 DOI: 10.1111/iej.13306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 04/06/2020] [Indexed: 11/27/2022]
Abstract
AIM To investigate the effect of ageing on control mechanisms of pulpal microcirculation using wavelet analysis and to calculate linear and nonlinear parameters of blood flow oscillations, in a healthy general population. METHODOLOGY Pulpal blood flow (PBF) oscillations were recorded on right maxillary central incisors using laser Doppler Flowmeter (PeriFlux PF 5001, Perimed, Jarfalla, Sweden) on a group of 10 young participants (20-25 years) and a group of 10 older adults (60-70 years). In total, 20 recordings were obtained for at least 20 min (one recording on one tooth per subject). Using wavelet spectral analysis, the amplitude and power were calculated as a linear and Hurst exponent as a nonlinear parameter of PBF oscillations. Differences between the two groups were assessed with the independent Student t-test. RESULTS Mean PBF levels were significantly lower (P = 0.024) in older adults than in young participants. Relative amplitudes and powers corresponding to the myogenic (P = 0.046, P < 0.001, respectively) and neurogenic activity (P = 0.04, P = 0.01, respectively) were significantly higher, whereas values corresponding to the endothelial function (P = 0.04, P = 0.01, respectively) were significantly lower in older adults than in young participants. Hurst exponents of the total spectrum, myogenic and endothelial component (P < 0.001, P = 0.02, P < 0.001, respectively) of PBF oscillations were significantly lower in older adults in comparison to young participants. CONCLUSIONS At the level of pulpal microcirculation, ageing was associated with altered blood flow levels, the contribution of different control mechanisms to blood flow oscillations as well as the interaction of vascular smooth muscle and endothelium. Described changes of pulpal haemodynamics contribute to a better understanding of physiological behaviour and decreased adaptability of aged dental pulp to pathological stimuli.
Collapse
Affiliation(s)
- B Dzeletovic
- DentalNet Research Group, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - D J Stratimirovic
- Department of Biophysics, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - D Stojic
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| | - L J Djukic
- Department of Pharmacology in Dentistry, School of Dental Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
46
|
Nam MCY, Meneses AL, Byrne CD, Richman T, Quah JX, Bailey TG, Hickman I, Anstey C, Askew CD, Senior R, Stanton T, Russell AW, Greaves K. An Experimental Series Investigating the Effects of Hyperinsulinemic Euglycemia on Myocardial Blood Flow Reserve in Healthy Individuals and on Myocardial Perfusion Defect Size following ST-Segment Elevation Myocardial Infarction. J Am Soc Echocardiogr 2020; 33:868-877.e6. [PMID: 32247531 DOI: 10.1016/j.echo.2020.01.010] [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: 07/21/2019] [Revised: 01/12/2020] [Accepted: 01/12/2020] [Indexed: 11/16/2022]
Abstract
BACKGROUND Incomplete restoration of myocardial blood flow (MBF) is reported in up to 30% of ST-segment elevation myocardial infarction (STEMI) despite prompt mechanical revascularization. Experimental hyperinsulinemic euglycemia (HE) increases MBF reserve (MBFR). If fully exploited, this effect may also improve MBF to ischemic myocardium. Using insulin-dextrose infusions to induce HE, we conducted four experiments to determine (1) how insulin infusion duration, dose, and presence of insulin resistance affect MBFR response; and (2) the effect of an insulin-dextrose infusion given immediately following revascularization of STEMI on myocardial perfusion. METHODS The MBFR was determined using myocardial contrast echocardiography. Experiment 1 (insulin duration): 12 participants received an insulin-dextrose or saline infusion for 120 minutes. MBFR was measured at four time intervals during infusion. Experiment 2 (insulin dose): 22 participants received one of three insulin doses (0.5, 1.5, 3.0 mU/kg/minute) for 60 minutes. Baseline and 60-minute MBFRs were determined. Experiment 3 (insulin resistance): five metabolic syndrome and six type 2 diabetes (T2DM) participants received 1.5 mU/kg/minute of insulin-dextrose for 60 minutes. Baseline and 60-minute MBFRs were determined. Experiment 4 (STEMI): following revascularization for STEMI, 20 patients were randomized to receive either 1.5 mU/kg/minute insulin-dextrose infusion for 120 minutes or standard care. Myocardial contrast echocardiography was performed at four time intervals to quantify percentage contrast defect length. RESULTS Experiment 1: MBFR increased with time through to 120 minutes in the insulin-dextrose group and did not change in controls. Experiment 2: compared with baseline, MBFR increased in the 1.5 (2.42 ± 0.39 to 3.25 ± 0.77, P = .002), did not change in the 0.5, and decreased in the 3.0 (2.64 ± 0.25 to 2.16 ± 0.33, P = .02) mU/kg/minute groups. Experiment 3: compared with baseline, MBFR increase was only borderline significant in metabolic syndrome and T2DM participants (1.98 ± 0.33 to 2.59 ± 0.45, P = .04, and 1.67 ± 0.35 to 2.14 ± 0.21, P = .05). Experiment 4: baseline percentage contrast defect length was similar in both groups but with insulin decreased with time and was significantly lower than in controls at 60 minutes (2.8 ± 5.7 vs 13.7 ± 10.6, P = .02). CONCLUSIONS Presence of T2DM, insulin infusion duration, and dose are important determinants of the MBFR response to HE. When given immediately following revascularization for STEMI, insulin-dextrose reduces perfusion defect size at one hour. Hyperinsulinemic euglycemia may improve MBF following ischemia, but further studies are needed to clarify this.
Collapse
Affiliation(s)
- Michael C Y Nam
- Department of Cardiology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Annelise L Meneses
- VasoActive Research Group, School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Christopher D Byrne
- Nutrition and Metabolism, Institute for Developmental Sciences, University of Southampton, Southampton, United Kingdom; Southampton National Institute for Health Research Biomedical Research Centre, University Hospital Southampton, Southampton, United Kingdom
| | - Tuppence Richman
- Department of Cardiology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Jing Xian Quah
- Department of Cardiology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia
| | - Tom G Bailey
- VasoActive Research Group, School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Ingrid Hickman
- Diamantina Institute for Cancer, Immunology and Metabolic Medicine, Princess Alexandra Hospital, University of Queensland, Woolloongabba, Queensland, Australia
| | - Chris Anstey
- Department of Intensive Care, Sunshine Coast Hospital and Health Services and University of Queensland, Birtinya, Queensland, Australia
| | - Christopher D Askew
- Department of Cardiology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; VasoActive Research Group, School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, Queensland, Australia
| | - Roxy Senior
- Biomedical Research Unit, National Heart and Lung Institute, Imperial College London, Royal Brompton Hospital, London, United Kingdom
| | - Tony Stanton
- Department of Cardiology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; VasoActive Research Group, School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Anthony W Russell
- Department of Diabetes and Endocrinology, Princess Alexandra Hospital, Brisbane, Queensland, Australia; PA Southside Clinical Unit, Faculty of Medicine, University of Queensland, Woolloongabba, Queensland, Australia
| | - Kim Greaves
- Department of Cardiology, Sunshine Coast University Hospital, Birtinya, Queensland, Australia; VasoActive Research Group, School of Health and Sport Sciences, University of the Sunshine Coast, Sippy Downs, Queensland, Australia; School of Medicine, University of Queensland, Brisbane, Queensland, Australia.
| |
Collapse
|
47
|
Acute Effects of Whole-Body Vibration on Peripheral Blood Flow, Vibrotactile Perception and Balance in Older Adults. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17031069. [PMID: 32046205 PMCID: PMC7037406 DOI: 10.3390/ijerph17031069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Revised: 02/04/2020] [Accepted: 02/06/2020] [Indexed: 01/10/2023]
Abstract
Background: Non-invasive application of whole-body vibration (WBV) has the potential for inducing improvements in impaired peripheral circulation, cutaneous sensation and balance among older adults. However, relevant studies have frequently applied high magnitudes of vibration and show conflicting and inconclusive results. Therefore, we attempted to ascertain the acute responses in those parameters from exposure of thirty older subjects to WBV of three different magnitudes, defined according to ISO 2631-1 (1997). Methods: Each subject randomly underwent four sessions of intervention (three bouts of 1 min exposure with 1 min between-bout rests): WBV at 15, 20, or 25 Hz with a peak-to-peak displacement of 4 mm, or control condition. Results: Both during and after intervention, dorsal foot skin blood flow increased significantly under 20 and 25 Hz exposure conditions with greater responses under the latter condition, the magnitude of which slightly exceeded the recommended value. Plantar vibrotactile perception showed significant increases after WBV exposure with overall greater responses under higher frequencies of vibration. In contrast, no WBV-induced change in balance was observed. Conclusions: WBV at 20 Hz with a magnitude within the recommended limit can be effective in inducing enhancements in peripheral blood flow; however, the same magnitude of vibration seems insufficient in improving balance among older adults.
Collapse
|
48
|
Aykar SS, Reynolds DE, McNamara MC, Hashemi NN. Manufacturing of poly(ethylene glycol diacrylate)-based hollow microvessels using microfluidics. RSC Adv 2020; 10:4095-4102. [PMID: 35492659 PMCID: PMC9049053 DOI: 10.1039/c9ra10264g] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022] Open
Abstract
Biocompatible and self-standing poly(ethylene glycol diacrylate)-based hollow microvessels were fabricated from a microfluidic device using microfluidic principles.
Collapse
Affiliation(s)
- Saurabh S. Aykar
- Department of Mechanical Engineering
- Iowa State University
- Ames
- USA
| | | | | | - Nicole N. Hashemi
- Department of Mechanical Engineering
- Iowa State University
- Ames
- USA
- Department of Biomedical Sciences
| |
Collapse
|
49
|
Sawicka D, Maciak S, Kozłowska H, Kasacka I, Kloza M, Sadowska A, Sokołowska E, Konarzewski M, Car H. Functional and structural changes in aorta of mice divergently selected for basal metabolic rate. J Comp Physiol B 2019; 190:101-112. [PMID: 31873784 DOI: 10.1007/s00360-019-01252-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 11/29/2019] [Accepted: 12/08/2019] [Indexed: 12/20/2022]
Abstract
Cardiovascular diseases (CVD) are one of the most common causes of mortality likely genetically linked to the variation in basal metabolic rate (BMR). A robust test of the significance of such association may be provided by artificial selection experiments on animals selected for diversification of BMR. Here we asked whether genetically determined differences in BMR correlate with anatomical shift in endothelium structure and if so, the relaxation and contraction responses of the aorta in mice from two lines of Swiss-Webster laboratory mice (Mus musculus) divergently selected for high or low BMR (HBMR and LBMR lines, respectively). Functional and structural study of aorta showed that a selection for divergent BMR resulted in the between-line difference in diastolic aortic capacity. The relaxation was stronger in aorta of the HBMR mice, which may stem from greater flexibility of aorta mediated by higher activity of Ca2+-activated K+ channels. Structural examination also indicated that HBMR mice had significantly thicker aorta's middle layer compared to LBMR animals. Such changes may promote arterial stiffness predisposing to cardiovascular diseases. BMR-related differences in the structure and relaxation ability of aortas in studied animals may be reminiscent of potential risk factors in the development of CVD in humans.
Collapse
Affiliation(s)
- Diana Sawicka
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295, Białystok, Poland.
| | - Sebastian Maciak
- Department of Evolutionary and Physiological Ecology, Faculty of Biology, University of Bialystok, ul. Ciołkowskiego 1J, 15-245, Białystok, Poland
| | - Hanna Kozłowska
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, ul. Mickiewicza 2A, 15-089, Białystok, Poland
| | - Irena Kasacka
- Department of Histology and Cytophysiology, Medical University of Bialystok, ul. Mickiewicza 2C, 15-222, Białystok, Poland
| | - Monika Kloza
- Department of Experimental Physiology and Pathophysiology, Medical University of Bialystok, ul. Mickiewicza 2A, 15-089, Białystok, Poland
| | - Anna Sadowska
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295, Białystok, Poland
| | - Emilia Sokołowska
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295, Białystok, Poland
| | - Marek Konarzewski
- Department of Evolutionary and Physiological Ecology, Faculty of Biology, University of Bialystok, ul. Ciołkowskiego 1J, 15-245, Białystok, Poland
| | - Halina Car
- Department of Experimental Pharmacology, Medical University of Bialystok, ul. Szpitalna 37, 15-295, Białystok, Poland
| |
Collapse
|
50
|
Abstract
The microcirculation maintains tissue homeostasis through local regulation of blood flow and oxygen delivery. Perturbations in microvascular function are characteristic of several diseases and may be early indicators of pathological changes in the cardiovascular system and in parenchymal tissue function. These changes are often mediated by various reactive oxygen species and linked to disruptions in pathways such as vasodilation or angiogenesis. This overview compiles recent advances relating to redox regulation of the microcirculation by adopting both cellular and functional perspectives. Findings from a variety of vascular beds and models are integrated to describe common effects of different reactive species on microvascular function. Gaps in understanding and areas for further research are outlined. © 2020 American Physiological Society. Compr Physiol 10:229-260, 2020.
Collapse
Affiliation(s)
- Andrew O Kadlec
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - David D Gutterman
- Department of Physiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Department of Medicine-Division of Cardiology, Medical College of Wisconsin, Milwaukee, Wisconsin, USA.,Cardiovascular Center, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|