1
|
Queiroz M, Sena CM. Perivascular adipose tissue: a central player in the triad of diabetes, obesity, and cardiovascular health. Cardiovasc Diabetol 2024; 23:455. [PMID: 39732729 PMCID: PMC11682657 DOI: 10.1186/s12933-024-02549-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/17/2024] [Indexed: 12/30/2024] Open
Abstract
Perivascular adipose tissue (PVAT) is a dynamic tissue that affects vascular function and cardiovascular health. The connection between PVAT, the immune system, obesity, and vascular disease is complex and plays a pivotal role in the pathogenesis of vascular diseases such as atherosclerosis, hypertension, and vascular inflammation. In cardiometabolic diseases, PVAT becomes a significant source of proflammatory adipokines, leading to increased infiltration of immune cells, in cardiometabolic diseases, PVAT becomes a significant source of proinflammatory adipokines, leading to increased infiltration of immune cells, promoting vascular smooth muscle cell proliferation and migrationpromoting vascular smooth muscle cell proliferation and migration. This exacerbates vascular dysfunction by impairing endothelial cell function and promoting endothelial activation. Dysregulated PVAT also contributes to hemodynamic alterations and hypertension through enhanced sympathetic nervous system activity and impaired vasodilatory capacity of PVAT-derived factors. Therapeutic interventions targeting key components of this interaction, such as modulating PVAT inflammation, restoring adipokine balance, and attenuating immune cell activation, hold promise for mitigating obesity-related vascular complications. Lifestyle interventions, pharmacological agents targeting inflammatory pathways, and surgical approaches aimed at reducing PVAT mass or improving adipose tissue function are potential therapeutic avenues for managing vascular diseases associated with obesity and PVAT dysfunction.
Collapse
Affiliation(s)
- Marcelo Queiroz
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-548, Coimbra, Portugal
| | - Cristina M Sena
- Institute of Physiology, iCBR, Faculty of Medicine, University of Coimbra, Subunit 1, polo 3, Azinhaga de Santa Comba, Celas, 3000-548, Coimbra, Portugal.
| |
Collapse
|
2
|
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
|
3
|
Carlini NA, Harber MP, Fleenor BS. Lower estimates of myocardial perfusion are associated with greater aortic perivascular adipose tissue density in humans independent of aortic stiffness. Am J Physiol Heart Circ Physiol 2024; 327:H927-H934. [PMID: 39150391 PMCID: PMC11482261 DOI: 10.1152/ajpheart.00436.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 08/12/2024] [Accepted: 08/12/2024] [Indexed: 08/17/2024]
Abstract
Aortic perivascular adipose tissue (aPVAT) density is associated with age-related aortic stiffness in humans and therefore, may contribute to cardiovascular dysfunction. A lower subendocardial viability ratio (SEVR), an estimate of myocardial perfusion, indicates greater cardiovascular disease (CVD) risk and is associated with aortic stiffness in clinical populations. However, the influence of aortic stiffness on the relation between aPVAT density and SEVR/cardiovascular (CV) hemodynamics in apparently healthy adults is unknown. We hypothesize that greater aPVAT density will be associated with lower SEVR and higher CV hemodynamics independent of aortic stiffness. Fourteen (6 males/8 females; mean age, 55.4 ± 5.6 yr; body mass index, 25.5 ± 0.6 kg/m2) adults completed resting measures of myocardial perfusion (SEVR), CV hemodynamics (pulse wave analysis), aortic stiffness [carotid-femoral pulse wave velocity (cfPWV)], and a computed tomography scan to acquire aPVAT and visceral adipose tissue (VAT) density. Greater aPVAT density (i.e., higher density) was associated with lower SEVR (r = -0.78, P < 0.001) and a higher systolic pressure time integral (r = 0.49, P = 0.03), forward pulse height (r = 0.49, P = 0.03), reflected pulse height (r = 0.55, P = 0.02), ejection duration (r = 0.56, P = 0.02), and augmentation pressure (r = 0.69, P = 0.003), but not with the diastolic pressure time integral (r = -0.22, P = 0.22). VAT density was not associated with SEVR or any CV hemodynamic endpoints (all, P > 0.05). Furthermore, the relation between aPVAT density and SEVR remained after adjusting for aortic stiffness (r = -0.66, P = 0.01) but not age (r = -0.24, P > 0.05). These data provide initial evidence for aPVAT as a novel yet understudied local fat depot contributing to lower myocardial perfusion in apparently healthy adults with aging.NEW & NOTEWORTHY Aortic perivascular adipose tissue (aPVAT) density is associated with aging and aortic stiffness in humans and, therefore, may contribute to lower myocardial perfusion. We demonstrate that greater aPVAT, but not visceral adipose tissue density is associated with lower myocardial perfusion and augmentation pressure independent of aortic stiffness, but not independent of age. These data provide novel evidence for aPVAT as a potential therapeutic target to improve myocardial perfusion and cardiovascular function in humans with aging.
Collapse
Affiliation(s)
- Nicholas A Carlini
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Matthew P Harber
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Bradley S Fleenor
- Clinical Exercise Physiology, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
- DeBusk College of Osteopathic Medicine, Lincoln Memorial University, Harrogate, Tennessee, United States
| |
Collapse
|
4
|
Osikoya O, Hula N, da Silva RDNO, Goulopoulou S. Perivascular Adipose Tissue and Uterine Artery Adaptations to Pregnancy. Microcirculation 2024; 31:e12857. [PMID: 38826057 DOI: 10.1111/micc.12857] [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: 10/02/2023] [Revised: 03/28/2024] [Accepted: 04/22/2024] [Indexed: 06/04/2024]
Abstract
Pregnancy is characterized by longitudinal maternal, physiological adaptations to support the development of a fetus. One of the cardinal maternal adaptations during a healthy pregnancy is a progressive increase in uterine artery blood flow. This facilitates sufficient blood supply for the development of the placenta and the growing fetus. Regional hemodynamic changes in the uterine circulation, such as a vast reduction in uterine artery resistance, are mainly facilitated by changes in uterine artery reactivity and myogenic tone along with remodeling of the uterine arteries. These regional changes in vascular reactivity have been attributed to pregnancy-induced adaptations of cell-to-cell communication mechanisms, with an emphasis on the interaction between endothelial and vascular smooth muscle cells. Perivascular adipose tissue (PVAT) is considered the fourth layer of the vascular wall and contributes to the regulation of vascular reactivity in most vascular beds and most species. This review focuses on mechanisms of uterine artery reactivity and the role of PVAT in pregnancy-induced maternal vascular adaptations, with an emphasis on the uterine circulation.
Collapse
Affiliation(s)
- Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center at Fort Worth, Fort Worth, Texas, USA
| | - Nataliia Hula
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, USA
| | - Renée de Nazaré Oliveira da Silva
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, USA
| | - Styliani Goulopoulou
- Lawrence D. Longo, MD Center for Perinatal Biology, Department of Basic Sciences, Loma Linda University, Loma Linda, California, USA
- Department of Gynecology and Obstetrics, Loma Linda University, Loma Linda, California, USA
| |
Collapse
|
5
|
Wang Y, Wang X, Chen Y, Zhang Y, Zhen X, Tao S, Dou J, Li P, Jiang G. Perivascular fat tissue and vascular aging: A sword and a shield. Pharmacol Res 2024; 203:107140. [PMID: 38513826 DOI: 10.1016/j.phrs.2024.107140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 02/16/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
The understanding of the function of perivascular adipose tissue (PVAT) in vascular aging has significantly changed due to the increasing amount of information regarding its biology. Adipose tissue surrounding blood vessels is increasingly recognized as a key regulator of vascular disorders. It has significant endocrine and paracrine effects on the vasculature and is mediated by the production of a variety of bioactive chemicals. It also participates in a number of pathological regulatory processes, including oxidative stress, immunological inflammation, lipid metabolism, vasoconstriction, and dilation. Mechanisms of homeostasis and interactions between cells at the local level tightly regulate the function and secretory repertoire of PVAT, which can become dysregulated during vascular aging. The PVAT secretion group changes from being reducing inflammation and lowering cholesterol to increasing inflammation and increasing cholesterol in response to systemic or local inflammation and insulin resistance. In addition, the interaction between the PVAT and the vasculature is reciprocal, and the biological processes of PVAT are directly influenced by the pertinent indicators of vascular aging. The architectural and biological traits of PVAT, the molecular mechanism of crosstalk between PVAT and vascular aging, and the clinical correlation of vascular age-related disorders are all summarized in this review. In addition, this paper aims to elucidate and evaluate the potential benefits of therapeutically targeting PVAT in the context of mitigating vascular aging. Furthermore, it will discuss the latest advancements in technology used for targeting PVAT.
Collapse
Affiliation(s)
- Yan Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianmin Wang
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Yang Chen
- School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China
| | - Yuelin Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Xianjie Zhen
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Siyu Tao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Jinfang Dou
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Peng Li
- Xinjiang Uygur Autonomous Region Hospital of Traditional Chinese Medicine, Xinjiang 830000, China
| | - Guangjian Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing 100029, China; School of Traditional Chinese Medicine, Xinjiang Medical University, Xinjiang 830011, China.
| |
Collapse
|
6
|
Thompson JM, Watts SW, Terrian L, Contreras GA, Rockwell C, Rendon CJ, Wabel E, Lockwood L, Bhattacharya S, Nault R. A cell atlas of thoracic aortic perivascular adipose tissue: a focus on mechanotransducers. Am J Physiol Heart Circ Physiol 2024; 326:H1252-H1265. [PMID: 38517229 PMCID: PMC11380965 DOI: 10.1152/ajpheart.00040.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 03/05/2024] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
Perivascular adipose tissue (PVAT) is increasingly recognized for its function in mechanotransduction. However, major gaps remain in our understanding of the cells present in PVAT, as well as how different cells contribute to mechanotransduction. We hypothesized that snRNA-seq would reveal the expression of mechanotransducers, and test one (PIEZO1) to illustrate the expression and functional agreement between single-nuclei RNA sequencing (snRNA-seq) and physiological measurements. To contrast two brown tissues, subscapular brown adipose tissue (BAT) was also examined. We used snRNA-seq of the thoracic aorta PVAT (taPVAT) and BAT from male Dahl salt-sensitive (Dahl SS) rats to investigate cell-specific expression mechanotransducers. Localization and function of the mechanostransducer PIEZO1 were further examined using immunohistochemistry (IHC) and RNAscope, as well as pharmacological antagonism. Approximately 30,000 nuclei from taPVAT and BAT each were characterized by snRNA-seq, identifying eight major cell types expected and one unexpected (nuclei with oligodendrocyte marker genes). Cell-specific differential gene expression analysis between taPVAT and BAT identified up to 511 genes (adipocytes) with many (≥20%) being unique to individual cell types. Piezo1 was the most highly, widely expressed mechanotransducer. The presence of PIEZO1 in the PVAT but not the adventitia was confirmed by RNAscope and IHC in male and female rats. Importantly, antagonism of PIEZO1 by GsMTX4 impaired the PVAT's ability to hold tension. Collectively, the cell compositions of taPVAT and BAT are highly similar, and PIEZO1 is likely a mechanotransducer in taPVAT.NEW & NOTEWORTHY This study describes the atlas of cells in the thoracic aorta perivascular adipose tissue (taPVAT) of the Dahl-SS rat, an important hypertension model. We show that mechanotransducers are widely expressed in these cells. Moreover, PIEZO1 expression is shown to be restricted to the taPVAT and is functionally implicated in stress relaxation. These data will serve as the foundation for future studies investigating the role of taPVAT in this model of hypertensive disease.
Collapse
Affiliation(s)
- Janice M Thompson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Leah Terrian
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, United States
| | - G Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, United States
| | - Cheryl Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - C Javier Rendon
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, Michigan, United States
| | - Emma Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Lizbeth Lockwood
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Sudin Bhattacharya
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Rance Nault
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
7
|
Thompson JM, Watts SW, Terrian L, Contreras GA, Rockwell C, Rendon CJ, Wabel E, Lockwood L, Bhattacharya S, Nault R. A Cell Atlas of Thoracic Aortic Perivascular Adipose Tissue: a focus on mechanotransducers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.09.561581. [PMID: 37873456 PMCID: PMC10592719 DOI: 10.1101/2023.10.09.561581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Perivascular adipose tissue (PVAT) is increasingly recognized for its function in mechanotransduction. To examine the cell-specificity of recognized mechanotransducers we used single nuclei RNA sequencing (snRNAseq) of the thoracic aorta PVAT (taPVAT) from male Dahl SS rats compared to subscapular brown adipose tissue (BAT). Approximately 30,000 nuclei from taPVAT and BAT each were characterized by snRNAseq, identifying 8 major cell types expected and one unexpected (nuclei with oligodendrocyte marker genes). Cell-specific differential gene expression analysis between taPVAT and BAT identified up to 511 genes (adipocytes) with many (≥20%) being unique to individual cell types. Piezo1 was the most highly, widely expressed mechanotransducer. Presence of PIEZO1 in the PVAT was confirmed by RNAscope® and IHC; antagonism of PIEZO1 impaired the PVAT's ability to hold tension. Collectively, the cell compositions of taPVAT and BAT are highly similar, and PIEZO1 is likely a mechanotransducer in taPVAT.
Collapse
Affiliation(s)
- Janice M. Thompson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - Stephanie W. Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - Leah Terrian
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - G. Andres Contreras
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Cheryl Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - C. Javier Rendon
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI, USA
| | - Emma Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Lizabeth Lockwood
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Sudin Bhattacharya
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Rance Nault
- Department of Biochemistry & Molecular Biology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
- Denotes lead contact
| |
Collapse
|
8
|
Onofrei VA, Anisie E, Zamfir CL, Ceasovschih A, Constantin M, Mitu F, Grigorescu ED, Petroaie AD, Timofte DV. Role of Chemerin and Perivascular Adipose Tissue Characteristics on Cardiovascular Risk Assessment by Arterial Stiffness Markers in Patients with Morbid Obesity. J Clin Med 2023; 12:jcm12082885. [PMID: 37109222 PMCID: PMC10145532 DOI: 10.3390/jcm12082885] [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: 02/12/2023] [Revised: 04/09/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVE The development of arterial stiffness (AS) in obesity is a multifactorial and complex process. The pleomorphic actions of adipokines and their local activity in perivascular adipose tissue (PVAT) are potential modulators of AS appearance and progression. We aimed to assess the correlations between two adipokines (chemerin, adiponectin), PVAT morphological changes (adipocyte size, blood vessel wall thickness) and AS parameters in the special subgroup of patients with morbid obesity. MATERIAL AND METHODS We enrolled 25 patients with morbid obesity and 25 non-obese patients, who were age- and gender-matched, untreated for cardiovascular risk factors, and admitted to hospital for laparoscopic surgical procedures (bariatric surgery for morbid obesity and non-inflammatory benign pathology surgery for non-obese patients). Before the surgical procedures, we evaluated demographic and anthropometric data and biochemical parameters including the studied adipokines. Arterial stiffness was evaluated using a Medexpert ArteriographTM TL2 device. In both groups, adipocyte size and vascular wall thickness as well as local adiponectin activity were analyzed in PVAT from intraoperative biopsies. RESULTS In our study, adiponectin (p = 0.0003), chemerin (p = 0.0001) and their ratio (p = 0.005) had statistically significant higher mean values in patients with morbid obesity compared to normal-weight patients. In patients with morbid obesity there were significant correlations between chemerin and AS parameters such as aortic pulse wave velocity (p = 0.006) and subendocardial viability index (p = 0.009). In the same group adipocyte size was significantly correlated with another AS parameter, namely, aortic systolic blood pressure (p = 0.030). In normal-weight patients, blood vessel wall thickness positively correlated with AS parameters such as brachial (p = 0.023) and aortic augmentation index (p = 0.023). An important finding was the negative adipoR1 and adipoR2 immunoexpression in PVAT adipocytes of patients with morbid obesity. Additionally, we found significant correlations between blood vessel wall thickness and blood fasting glucose (p < 0.05) in both groups. CONCLUSIONS Chemerin and adipocyte size could be predictive biomarkers for AS in patients with morbid obesity. Given the small number of patients included, our results need further validation.
Collapse
Affiliation(s)
- Viviana Aursulesei Onofrei
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon, Clinical Emergency Hospital, Independence Boulevard No. 1, 700111 Iasi, Romania
| | - Ecaterina Anisie
- Cardiology Clinic, St. Spiridon, Clinical Emergency Hospital, Independence Boulevard No. 1, 700111 Iasi, Romania
| | - Carmen Lacramioara Zamfir
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
| | - Alexandr Ceasovschih
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon, Clinical Emergency Hospital, Independence Boulevard No. 1, 700111 Iasi, Romania
| | - Mihai Constantin
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
| | - Florin Mitu
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
- Clinical Rehabilitation Hospital, Cardiovascular Rehabilitation Clinic, Pantelimon Halipa Street No. 14, 700661 Iasi, Romania
- Academy of Medical Sciences, Ion C. Brătianu Boulevard No. 1, 030173 Bucharest, Romania
- Romanian Academy of Scientists, Dimitrie Mangeron Boulevard No. 433, 700050 Iasi, Romania
| | - Elena-Daniela Grigorescu
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
| | - Antoneta Dacia Petroaie
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
| | - Daniel Vasile Timofte
- Department of Medical Specialties, Grigore T. Popa, University of Medicine and Pharmacy, University Street No. 16, 700115 Iasi, Romania
- Cardiology Clinic, St. Spiridon, Clinical Emergency Hospital, Independence Boulevard No. 1, 700111 Iasi, Romania
- Academy of Medical Sciences, Ion C. Brătianu Boulevard No. 1, 030173 Bucharest, Romania
| |
Collapse
|
9
|
Cheng CK, Ding H, Jiang M, Yin H, Gollasch M, Huang Y. Perivascular adipose tissue: Fine-tuner of vascular redox status and inflammation. Redox Biol 2023; 62:102683. [PMID: 36958248 PMCID: PMC10038789 DOI: 10.1016/j.redox.2023.102683] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/14/2023] [Accepted: 03/17/2023] [Indexed: 03/22/2023] Open
Abstract
Perivascular adipose tissue (PVAT) refers to the aggregate of adipose tissue surrounding the vasculature, exhibiting the phenotypes of white, beige and brown adipocytes. PVAT has emerged as an active modulator of vascular homeostasis and pathogenesis of cardiovascular diseases in addition to its structural role to provide mechanical support to blood vessels. More specifically, PVAT is closely involved in the regulation of reactive oxygen species (ROS) homeostasis and inflammation along the vascular tree, through the tight interaction between PVAT and cellular components of the vascular wall. Furthermore, the phenotype-genotype of PVAT at different regions of vasculature varies corresponding to different cardiovascular risks. During ageing and obesity, the cellular proportions and signaling pathways of PVAT vary in favor of cardiovascular pathogenesis by promoting ROS generation and inflammation. Physiological means and drugs that alter PVAT mass, components and signaling may provide new therapeutic insights in the treatment of cardiovascular diseases. In this review, we aim to provide an updated understanding towards PVAT in the context of redox regulation, and to highlight the therapeutic potential of targeting PVAT against cardiovascular complications.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| | - Huanyu Ding
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Minchun Jiang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Huiyong Yin
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Felix-Hausdorff-Straße 3, 17487, Greifswald, Germany
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
10
|
Fleenor BS, Carlini NA, Ouyang A, Du B, Harber MP. Greater aortic perivascular adipose tissue density is associated with aging, aortic stiffness, and central blood pressure in humans. J Appl Physiol (1985) 2023; 134:703-709. [PMID: 36759156 DOI: 10.1152/japplphysiol.00745.2022] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 01/17/2023] [Accepted: 02/01/2023] [Indexed: 02/11/2023] Open
Abstract
Aging results in aortic perivascular adipose tissue (aPVAT)-mediated aortic stiffening in preclinical animal models to promote cardiovascular dysfunction. We hypothesized that greater human aPVAT density will be associated with aging, higher aortic stiffness, and blood pressure (BP). Fourteen apparently healthy adults (6 M/8 F, age range 20-79 yr) were recruited for this study. Aortic stiffness, assessed by carotid-femoral pulse wave velocity (cfPWV), resting aortic BP via pulse wave analysis, and aPVAT and abdominal visceral adipose tissue (VAT) density by computed tomography attenuation were acquired. aPVAT and epididymal (visceral) fat from young (4-6 mo) and old (27-29 mo) mice were used for ex vivo-conditioned media intrinsic mechanical stiffness experiments. Compared with younger adults, older adults had higher cfPWV (8.6 ± 0.4 vs. 6.2 ± 0.6 m/s, P < 0.05) and greater aPVAT attenuation (-80.2 ± 2.0 vs. -95.9 ± 1.5 HU, P < 0.05), but not VAT attenuation (P > 0.05). aPVAT-conditioned media from old mice compared with young mice increased intrinsic mechanical stiffness of the aorta (4,519 ± 510 vs. 2,325 ± 563 kPa, P < 0.05), which was not observed with epididymal fat-conditioned media from old mice (P > 0.05). aPVAT, but not VAT density, was positively associated with age (r = 0.89), cfPWV (r = 0.56), resting augmentation index normalized to heart rate 75 (AIxHR75; r = 0.67), aortic systolic BP (r = 0.58), and aortic pulse pressure (PP; r = 0.59; P < 0.05, all) and were independent of VAT density (P < 0.05, all). These data herein provide evidence for aPVAT as a novel fat depot and therapeutic target to lower aortic stiffness and future cardiovascular disease risk with aging in humans.NEW & NOTEWORTHY Aortic perivascular adipose tissue (aPVAT) promotes age-related aortic stiffening in preclinical animal models, but the relation between aPVAT density and cardiovascular function in adults is unknown. We demonstrate that aPVAT, but not abdominal visceral adipose tissue density, is positively associated with aging, aortic stiffness, and higher resting aortic blood pressure in apparently healthy adults. These findings provide novel evidence for aPVAT as a viable therapeutic target for improving cardiovascular function in humans.
Collapse
Affiliation(s)
- Bradley S Fleenor
- Clinical Exercise Physiology Program, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - Nicholas A Carlini
- Clinical Exercise Physiology Program, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| | - An Ouyang
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States
| | - Bing Du
- Department of Cardiology, First Hospital of Jilin University, Changchun, People's Republic of China
| | - Matthew P Harber
- Clinical Exercise Physiology Program, Human Performance Laboratory, Ball State University, Muncie, Indiana, United States
| |
Collapse
|
11
|
Aerobic Exercise Prevents Arterial Stiffness and Attenuates Hyperexcitation of Sympathetic Nerves in Perivascular Adipose Tissue of Mice after Transverse Aortic Constriction. Int J Mol Sci 2022; 23:ijms231911189. [PMID: 36232489 PMCID: PMC9570255 DOI: 10.3390/ijms231911189] [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: 09/07/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/17/2022] Open
Abstract
We aimed to investigate the efficacy of exercise on preventing arterial stiffness and the potential role of sympathetic nerves within perivascular adipose tissue (PVAT) in pressure-overload-induced heart failure (HF) mice. Eight-week-old male mice were subjected to sham operation (SHAM), transverse aortic constriction-sedentary (TAC-SE), and transverse aortic constriction-exercise (TAC-EX) groups. Six weeks of aerobic exercise training was performed using a treadmill. Arterial stiffness was determined by measuring the elastic modulus. The elastic and collagen fibers of the aorta and sympathetic nerve distribution in PVAT were observed. Circulating noradrenaline (NE), expressions of β3-adrenergic receptor (β3-AR), and adiponectin in PVAT were quantified. During the recovery of cardiac function by aerobic exercise, thoracic aortic collagen elastic modulus (CEM) and collagen fibers were significantly decreased (p < 0.05, TAC-SE vs. TAC-EX), and elastin elastic modulus (EEM) was significantly increased (p < 0.05, TAC-SE vs. TAC-EX). Circulating NE and sympathetic nerve distribution in PVAT were significantly decreased (p < 0.05, TAC-SE vs. TAC-EX). The expression of β3-AR was significantly reduced (p < 0.05, TAC-SE vs. TAC-EX), and adiponectin was significantly increased (p < 0.05, TAC-SE vs. TAC-EX) in PVAT. Regular aerobic exercise can effectively prevent arterial stiffness and extracellular matrix (ECM) remodeling in the developmental course of HF, during which sympathetic innervation and adiponectin within PVAT might be strongly implicated.
Collapse
|
12
|
Perivascular adipose tissue-mediated arterial stiffening in aging and disease: An emerging translational therapeutic target? Pharmacol Res 2022; 178:106150. [PMID: 35339679 DOI: 10.1016/j.phrs.2022.106150] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Revised: 02/14/2022] [Accepted: 02/26/2022] [Indexed: 01/11/2023]
Abstract
Cardiovascular diseases (CVD) are the leading cause of mortality in modernized societies. Arterial stiffening with aging and disease is a key pathological event leading to increased CVD morbidity and mortality. Perivascular adipose tissue (PVAT) is a fat depot not widely studied yet has direct and profound effects on arterial stiffening. Identifying PVAT as a novel therapeutic target to lower arterial stiffness and thereby CVD risk has potentially important clinical ramifications. Thus, herein, we will overview the current preclinical evidence and the associated mechanisms for PVAT to promote arterial stiffness with aging and other disease conditions. We will also discuss viable translational lifestyle and pharmacological interventions for altering PVAT function that may de-stiffen arteries. Last, the translational potential for PVAT as a therapeutic target to lower arterial stiffness and CVD risk for clinical populations will be discussed.
Collapse
|
13
|
Barros PR, Costa TJ, Akamine EH, Tostes RC. Vascular Aging in Rodent Models: Contrasting Mechanisms Driving the Female and Male Vascular Senescence. FRONTIERS IN AGING 2021; 2:727604. [PMID: 35821995 PMCID: PMC9261394 DOI: 10.3389/fragi.2021.727604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Increasing scientific interest has been directed to sex as a biological and decisive factor on several diseases. Several different mechanisms orchestrate vascular function, as well as vascular dysfunction in cardiovascular and metabolic diseases in males and females. Certain vascular sex differences are present throughout life, while others are more evident before the menopause, suggesting two important and correlated drivers: genetic and hormonal factors. With the increasing life expectancy and aging population, studies on aging-related diseases and aging-related physiological changes have steeply grown and, with them, the use of aging animal models. Mouse and rat models of aging, the most studied laboratory animals in aging research, exhibit sex differences in many systems and physiological functions, as well as sex differences in the aging process and aging-associated cardiovascular changes. In the present review, we introduce the most common aging and senescence-accelerated animal models and emphasize that sex is a biological variable that should be considered in aging studies. Sex differences in the cardiovascular system, with a focus on sex differences in aging-associated vascular alterations (endothelial dysfunction, remodeling and oxidative and inflammatory processes) in these animal models are reviewed and discussed.
Collapse
Affiliation(s)
- Paula R. Barros
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tiago J. Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eliana H. Akamine
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
- *Correspondence: Rita C. Tostes, ; Eliana H. Akamine,
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Rita C. Tostes, ; Eliana H. Akamine,
| |
Collapse
|
14
|
van der Bruggen MM, Spronck B, Delhaas T, Reesink KD, Schalkwijk CG. The Putative Role of Methylglyoxal in Arterial Stiffening: A Review. Heart Lung Circ 2021; 30:1681-1693. [PMID: 34393049 DOI: 10.1016/j.hlc.2021.06.527] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/28/2021] [Accepted: 06/01/2021] [Indexed: 12/13/2022]
Abstract
BACKGROUND Arterial stiffening is a hallmark of vascular ageing and a consequence of many diseases including diabetes mellitus. Methylglyoxal (MGO), a highly reactive α-dicarbonyl mainly formed during glycolysis, has emerged as a potential contributor to the development of arterial stiffness. MGO reacts with arginine and lysine residues in proteins to form stable advanced glycation endproducts (AGEs). AGEs may contribute to arterial stiffening by increased cross-linking of collagen within the extracellular matrix (ECM), by altering the vascular structure, and by triggering inflammatory and oxidative pathways. Although arterial stiffness is mainly determined by ECM and vascular smooth muscle cell function, the effects of MGO and MGO-derived AGEs on these structures have not been thoroughly reviewed to date. METHODS AND RESULTS We conducted a PubMed search without filtering for publication date which resulted in 16 experimental and 22 clinical studies eligible for inclusion. Remarkably, none of the experimental and only three of the clinical studies specifically mentioned MGO-derived AGEs. Almost all studies reported an association between arterial stiffness and AGE accumulation in the arterial wall or increased plasma AGEs. Other studies report reduced arterial stiffness in experimental models upon administration of AGE-breakers. CONCLUSIONS No papers published to date directly show an association between MGO or MGO-derived AGEs and arterial stiffening. The relevance of the various underlying mechanisms is not yet clear, which is particularly due to methodological challenges in the detection of MGO and MGO-derived AGEs at the molecular, intra- and pericellular, and structural levels, as well as in challenges in the assessment of intrinsic arterial wall properties at ECM- and tissue levels.
Collapse
Affiliation(s)
- Myrthe M van der Bruggen
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Bart Spronck
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands; Department of Biomedical Engineering, School of Engineering & Applied Sciences, Yale University, New Haven, CT, USA
| | - Tammo Delhaas
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands
| | - Koen D Reesink
- Department of Biomedical Engineering, CARIM School for Cardiovascular Diseases, Maastricht University, Maastricht, The Netherlands.
| | - Casper G Schalkwijk
- Department of Internal Medicine, CARIM School for Cardiovascular Diseases, Maastricht University Medical Center, Maastricht, The Netherlands
| |
Collapse
|
15
|
Valls RM, Pedret A, Calderón-Pérez L, Llauradó E, Pla-Pagà L, Companys J, Moragas A, Martín-Luján F, Ortega Y, Giralt M, Romeu M, Rubió L, Mayneris-Perxachs J, Canela N, Puiggrós F, Caimari A, Del Bas JM, Arola L, Solà R. Effects of hesperidin in orange juice on blood and pulse pressures in mildly hypertensive individuals: a randomized controlled trial (Citrus study). Eur J Nutr 2020; 60:1277-1288. [PMID: 32661681 PMCID: PMC7987641 DOI: 10.1007/s00394-020-02279-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 05/11/2020] [Indexed: 12/25/2022]
Abstract
Purpose To assess the sustained and acute effects, as well as the influence of sustained consumption on the acute effects, of orange juice (OJ) with a natural hesperidin content and hesperidin-enriched OJ (EOJ) on blood (BP) and pulse (PP) pressures in pre- and stage-1 hypertensive individuals. Methods In a randomized, parallel, double-blind, placebo-controlled trial, participants (n = 159) received 500 mL/day of control drink, OJ, or EOJ for 12 weeks. Two dose–response studies were performed at baseline and after 12 weeks. Results A single EOJ dose (500 mL) reduced systolic BP (SBP) and PP, with greater changes after sustained treatment where a decrease in diastolic BP (DBP) also occurred (P < 0.05). SBP and PP decreased in a dose-dependent manner relative to the hesperidin content of the beverages throughout the 12 weeks (P < 0.05). OJ and EOJ decreased homocysteine levels at 12 weeks versus the control drink (P < 0.05). After 12 weeks of EOJ consumption, four genes related to hypertension (PTX3, NLRP3, NPSR1 and NAMPT) were differentially expressed in peripheral blood mononuclear cells (P < 0.05). Conclusion Hesperidin in OJ reduces SBP and PP after sustained consumption, and after a single dose, the chronic consumption of EOJ enhances its postprandial effect. Decreases in systemic and transcriptomic biomarkers were concomitant with BP and PP changes. EOJ could be a useful co-adjuvant tool for BP and PP management in pre- and stage-1 hypertensive individuals. Electronic supplementary material The online version of this article (10.1007/s00394-020-02279-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Rosa M Valls
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Anna Pedret
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain.
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain.
| | - Lorena Calderón-Pérez
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Elisabet Llauradó
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Laura Pla-Pagà
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Judit Companys
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Ana Moragas
- Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain
- Institut Universitari d'Investigació en Atenció Primària-IDIAP Jordi Gol, Tarragona, Spain
- Primary Care Centre Jaume I, Institut Català de la Salut, Tarragona, Spain
| | - Francisco Martín-Luján
- Departament de Medicina i Cirurgia, Universitat Rovira i Virgili, Reus, Spain
- Institut Universitari d'Investigació en Atenció Primària-IDIAP Jordi Gol, Tarragona, Spain
- Primary Care Centre El Morell, Institut Català de la Salut, Tarragona, Spain
| | - Yolanda Ortega
- Institut Universitari d'Investigació en Atenció Primària-IDIAP Jordi Gol, Tarragona, Spain
- Primary Care Centre Jaume I, Institut Català de la Salut, Tarragona, Spain
- Primary Care Centre Salou, Institut Català de la Salut, Tarragona, Spain
| | - Montse Giralt
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
| | - Marta Romeu
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
| | - Laura Rubió
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Food Technology Department, XaRTA-TPV, Agrotecnio Center, Escola Tècnica Superior d'Enginyeria Agrària, University of Lleida, Av/ Alcalde Rovira Roure 191, 25198, Lleida, Spain
| | - Jordi Mayneris-Perxachs
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences, Reus, Spain
| | - Núria Canela
- Eurecat, Centre Tecnològic de Catalunya, Centre for Omic Sciences, Reus, Spain
| | - Francesc Puiggrós
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Antoni Caimari
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Josep M Del Bas
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
| | - Lluís Arola
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
- Departament de Bioquímica i Biotecnologia, Grup de Recerca en Nutrigenòmica, Universitat Rovira i Virgili, Tarragona, Spain
| | - Rosa Solà
- Functional Nutrition, Oxidation, and Cardiovascular Diseases Group (NFOC-Salut), Universitat Rovira i Virgili, Facultat de Medicina i Ciències de La Salut, Reus, Spain
- Eurecat, Centre Tecnològic de Catalunya, Unitat de Nutrició i Salut, Av. de La Universitat, 1, 43204, Reus, Spain
- Hospital Universitari Sant Joan de Reus, Reus, Spain
| |
Collapse
|
16
|
Zheng M, Lu S, Xing J. Enhanced antioxidant, anti-inflammatory and α-glucosidase inhibitory activities of citrus hesperidin by acid-catalyzed hydrolysis. Food Chem 2020; 336:127539. [PMID: 32763730 DOI: 10.1016/j.foodchem.2020.127539] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 02/06/2023]
Abstract
Hesperidin hydrolysates (HHS) was produced by the hydrolysis of hesperidin (HDN) in previous studies. The potential components in HHS were identified by LC-MS, and minor components (MCS) in HHS were isolated. Antioxidant activities by radical-scavenging capacities, reducing capacity and β-carotene-linoleate assay, anti-inflammatory effects by inhibiting NO production of RAW 264.7 cells, and α-glucosidase inhibitory effects of HDN, HHS, MCS and henperetin (HTN) were investigated in present study. HHS showed higher radical scavenging activities, higher reducing capacity, and higher inhibitory activity in the β-carotene-linoleate assay than HDN. HHS inhibited the production of NO and pro-inflammatory cytokines of RAW 264.7 cells more strongly than HDN. HHS also intensively inhibited α-glucosidase activity whereas HDN showed little activity. In addition, the effects of MCS on above activities showed it play a synergistic part with HTN. This work suggested that hydrolyzation of HDN enhance the activities, and provided valuable information on effective utilization of HDN.
Collapse
Affiliation(s)
- Meiyu Zheng
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Key Laboratory of Fruit and Vegetables Postharvest and Processing Technology Research of Zhejiang Province, Key Laboratory of Postharvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Hangzhou 310021, China
| | - Shengmin Lu
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Key Laboratory of Fruit and Vegetables Postharvest and Processing Technology Research of Zhejiang Province, Key Laboratory of Postharvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Hangzhou 310021, China.
| | - Jianrong Xing
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Key Laboratory of Fruit and Vegetables Postharvest and Processing Technology Research of Zhejiang Province, Key Laboratory of Postharvest Handling of Fruits, Ministry of Agriculture and Rural Affairs, Hangzhou 310021, China
| |
Collapse
|
17
|
Mahanes TM, Murphy MO, Ouyang A, Yiannikouris FB, Fleenor BS, Loria AS. Maternal separation-induced increases in vascular stiffness are independent of circulating angiotensinogen levels. J Appl Physiol (1985) 2020; 129:58-65. [PMID: 32407243 DOI: 10.1152/japplphysiol.00703.2019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The renin-angiotensin system (RAS) precursor angiotensinogen (AGT) has been implicated in the functional and mechanical alterations of the vascular wall in response to high-fat diet (HFD). Previously, we showed that HFD exacerbates angiotensin II-induced constriction in isolated aortic rings from male rats exposed to maternal separation (MatSep), a model of early-life stress. Thus, the aim of this study was to investigate whether MatSep increases AGT secretion promoting vascular stiffness in rats fed a HFD. Male Wistar-Kyoto MatSep offspring were separated (3 h/day, postnatal days 2-14), and undisturbed littermates were used as controls. At weaning, rats were fed for 17 wk a normal diet (ND) or a HFD, 18% or 60% kcal from fat, respectively. In plasma, there was a main effect of MatSep reducing AGT concentration (P < 0.05) but no effect due to diet. In urine, ND-fed MatSep rats displayed higher AGT concentrations that were further increased by HFD (P < 0.05 vs. control). AGT mRNA abundance and protein expression were increased in adipose tissue from HFD-fed MatSep rats compared with control rats (P < 0.05). No significant differences in liver and kidney AGT levels were found between groups. In addition, MatSep augmented vascular stiffness assessed on freshly isolated aortic rings from ND-fed rats (P < 0.05), yet HFD did not worsen vascular stiffness in either MatSep or control rats. There was no correlation between plasma AGT and vascular stiffness in ND-fed rats; however, this relationship was negative in HFD-fed MatSep rats only (P < 0.05). Therefore, this study shows that MatSep-induced increases in vascular stiffness are independent of diet or plasma AGT.NEW & NOTEWORTHY This study demonstrates that there was no correlation between circulating levels of angiotensinogen (AGT) and the development of vascular stiffness in rats exposed to early-life stress and fed a normal diet. This study also shows that early-life stress-induced hypersensitive vascular contractility to angiotensin II in rats fed a high-fat diet is independent of circulating levels of AGT and occurs without further progression of vascular stiffness. Our data show that early-life stress primes the adipose tissue to secrete AGT in a sex- and species-independent fashion.
Collapse
Affiliation(s)
- Timothy M Mahanes
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - Margaret O Murphy
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| | - An Ouyang
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | | | | | - Analia S Loria
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, Lexington, Kentucky
| |
Collapse
|
18
|
Jack BU, Malherbe CJ, Mamushi M, Muller CJF, Joubert E, Louw J, Pheiffer C. Adipose tissue as a possible therapeutic target for polyphenols: A case for Cyclopia extracts as anti-obesity nutraceuticals. Biomed Pharmacother 2019; 120:109439. [PMID: 31590126 DOI: 10.1016/j.biopha.2019.109439] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 08/29/2019] [Accepted: 09/06/2019] [Indexed: 02/07/2023] Open
Abstract
Obesity is a significant contributor to increased morbidity and premature mortality due to increasing the risk of many chronic metabolic diseases such as type 2 diabetes, cardiovascular disease and certain types of cancer. Lifestyle modifications such as energy restriction and increased physical activity are highly effective first-line treatment strategies used in the management of obesity. However, adherence to these behavioral changes is poor, with an increased reliance on synthetic drugs, which unfortunately are plagued by adverse effects. The identification of new and safer anti-obesity agents is thus of significant interest. In recent years, plants and their phenolic constituents have attracted increased attention due to their health-promoting properties. Amongst these, Cyclopia, an endemic South African plant commonly consumed as a herbal tea (honeybush), has been shown to possess modulating properties against oxidative stress, hyperglycemia, and obesity. Likewise, several studies have reported that some of the major phenolic compounds present in Cyclopia spp. exhibit anti-obesity effects, particularly by targeting adipose tissue. These phenolic compounds belong to the xanthone, flavonoid and benzophenone classes. The aim of this review is to assess the potential of Cyclopia extracts as an anti-obesity nutraceutical as underpinned by in vitro and in vivo studies and the underlying cellular mechanisms and biological pathways regulated by their phenolic compounds.
Collapse
Affiliation(s)
- Babalwa U Jack
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa.
| | - Christiaan J Malherbe
- Plant Bioactives Group, Post-Harvest and Agro-processing Technologies, Agricultural Research Council, Infruitec-Nietvoorbij, Stellenbosch, South Africa
| | - Mokadi Mamushi
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| | - Christo J F Muller
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa; Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa, South Africa
| | - Elizabeth Joubert
- Plant Bioactives Group, Post-Harvest and Agro-processing Technologies, Agricultural Research Council, Infruitec-Nietvoorbij, Stellenbosch, South Africa; Department of Food Science, Stellenbosch University, Stellenbosch, South Africa
| | - Johan Louw
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Department of Biochemistry and Microbiology, University of Zululand, Kwa-Dlangezwa, South Africa
| | - Carmen Pheiffer
- Biomedical Research and Innovation Platform, South African Medical Research Council, Tygerberg, South Africa; Division of Medical Physiology, Faculty of Medicine and Health Sciences, Stellenbosch University, Tygerberg, South Africa
| |
Collapse
|
19
|
Ouyang A, Olver TD, Emter CA, Fleenor BS. Chronic exercise training prevents coronary artery stiffening in aortic-banded miniswine: role of perivascular adipose-derived advanced glycation end products. J Appl Physiol (1985) 2019; 127:816-827. [PMID: 31295062 DOI: 10.1152/japplphysiol.00146.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Heart failure (HF) is associated with increased large conduit artery stiffness and afterload resulting in stiffening of the coronary arteries. Perivascular adipose tissue (PVAT) and advanced glycation end products (AGE) both promote arterial stiffness, yet the mechanisms by which coronary PVAT promotes arterial stiffness and the efficacy of exercise to prevent coronary stiffness are unknown. We hypothesized that both chronic continuous and interval exercise training would prevent coronary PVAT-mediated AGE secretion and arterial stiffness. Yucatan miniature swine were divided into four groups: control-sedentary (CON), aortic banded sedentary-heart failure (HF), aortic banded HF-continuous exercise trained (HF+CONT), and aortic banded HF-interval exercise trained (HF+IT). The left circumflex and right coronary arteries underwent ex vivo mechanical testing, and arterial AGE, elastin, and collagen were assessed. Coronary elastin elastic modulus (EEM) and elastin protein were lower and AGE was increased with HF compared with CON, which was prevented by both HF+CONT and HF+IT. Mouse aortic segments treated with swine coronary PVAT conditioned medium had lower EEM and elastin content and greater AGE secretion and arterial AGE accumulation in HF compared with CON, which was prevented by both HF+CONT and HF+IT. Aminoguanidine (AMG), an AGE inhibitor, prevented the reduction in EEM, arterial elastin content, and AGE accumulation in mouse aortic segments treated with PVAT conditioned medium in the HF group. Our data demonstrate efficacy for chronic continuous and interval exercise to prevent coronary artery stiffness via inhibition of PVAT-derived AGE secretion in a preclinical miniswine model of pressure overload-induced HF.NEW & NOTEWORTHY Our findings show that chronic continuous and interval exercise training regimens prevent coronary artery stiffness associated with inhibition of perivascular adipose tissue-derived advanced glycation end products in a translational pressure overload-induced heart failure model potentially providing an effective therapeutic option for heart failure patients.
Collapse
Affiliation(s)
- An Ouyang
- Department of Kinesiology and Health Promotion, University of Kentucky, Lexington, Kentucky
| | - T Dylan Olver
- Department of Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Craig A Emter
- Department of Biomedical Sciences, University of Missouri-Columbia, Columbia, Missouri
| | - Bradley S Fleenor
- Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, Indiana
| |
Collapse
|
20
|
Zhang ZM, Wang BX, Ou WS, Lv YH, Li MM, Miao Z, Wang SX, Fei JC, Guo T. Administration of losartan improves aortic arterial stiffness and reduces the occurrence of acute coronary syndrome in aged patients with essential hypertension. J Cell Biochem 2018; 120:5713-5721. [PMID: 30362602 DOI: 10.1002/jcb.27856] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 09/19/2018] [Indexed: 02/03/2023]
Abstract
BACKGROUNDS AND AIMS Increased arterial stiffness may increase cardiovascular morbidity and mortality. Angiotensin II type 1 receptor blocker losartan is potentially useful in controlling the central blood pressure and arterial stiffness in mild to moderate essential hypertension, while the effects of losartan in aged patients with essential hypertension are not entirely investigated. METHODS The carotid-femoral arterial pulse wave velocity (PWV) was measured in aged patients with essential hypertension. RESULTS In a cross-sectional study, PWV value was significantly higher in these old patients with essential hypertension, compared with patients without essential hypertension. Logistic regression analysis indicated that age, hypertension duration, and losartan treatment are risk factors of arterial stiffness. In a perspective study, long-term administration of losartan (50 mg/d) remarkably reduced PWV in aged patients with essential hypertension. In a longitudinal study, PWV is an independent predictor of the occurrence of acute coronary syndrome (ACS) in elderly patients with essential hypertension by using multivariate analysis. Further, the ACS occurrence was reduced by long-term administration of losartan in aged patients with essential hypertension, compared with the old hypertensive patients without taking losartan. CONCLUSION Losartan treatment is a negative risk factor of arterial stiffness and reduces the risk of ACS in aged patients with essential hypertension.
Collapse
Affiliation(s)
- Zhi-Mian Zhang
- Department of Cardiology, The Center of Health Examination, Qilu Hospital, Shandong University, Jinan, China
| | - Bing-Xiang Wang
- Department of Orthopedics, Provincial Hospital of Shandong, Jinan, China
| | - Wen-Sheng Ou
- Department of Liver Disease, Chenzhou No.1 People s Hospital, Chenzhou, China
| | - Yan-Hong Lv
- Department of Cardiology, The Center of Health Examination, Qilu Hospital, Shandong University, Jinan, China
| | - Ming-Min Li
- Department of Cardiology, The Center of Health Examination, Qilu Hospital, Shandong University, Jinan, China
| | - Zhang Miao
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Shuang-Xi Wang
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| | - Jian-Chun Fei
- Department of Anaesthesia, Qilu Hospital, Shandong University, Jinan, China
| | - Tao Guo
- The Key Laboratory of Cardiovascular Remodeling and Function Research, The State and Shandong Province Joint Key Laboratory of Translational Cardiovascular Medicine, Department of Cardiology, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
21
|
Zhu ML, Sun RL, Zhang HY, Zhao FR, Pan GP, Zhang C, Song P, Li P, Xu J, Wang S, Yin YL. Angiotensin II type 1 receptor blockers prevent aortic arterial stiffness in elderly patients with hypertension. Clin Exp Hypertens 2018; 41:657-661. [PMID: 30311805 DOI: 10.1080/10641963.2018.1529781] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Backgrounds and aims: Increased arterial stiffness may increase cardiovascular morbidity and mortality. Angiotensin II type 1 receptor blockers (ARBs) are potentially useful in controlling the central blood pressure and arterial stiffness in mild to moderate essential hypertension, while the effects of ARBs in aged patients with essential hypertension are not entirely investigated. Methods: The carotid-femoral arterial pulse wave velocity (PWV) was measured in aged patients with essential hypertension. Results: In a cross-sectional study, PWV value was significantly higher in these old patients with essential hypertension, compared to patients without essential hypertension. In correlation analysis, PWV was associated positively with age, hypertension duration, and carotid atherosclerosis. However, there was no relationship between PWV and gender in aged patients with essential hypertension. In a perspective study, 6-12 months administration of ARBs (losartan, 50 mg/day; telmisartan, 40 mg/day; valsartan 80 mg/day; irbesartan, 150 mg/day) remarkably reduced PWV in aged patients with essential hypertension. Regression analyses of multiple factors indicated that the effects of ARBs on arterial stiffness were not associated with the reduction of blood pressure. Conclusion: ARB treatment is a negative risk factor of arterial stiffness in aged patients with essential hypertension.
Collapse
Affiliation(s)
- Mo-Li Zhu
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Rui-Li Sun
- b Henan Key Laboratory of Immunology and Targeted Therapy, Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University , Xinxiang , China
| | - He-Yun Zhang
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Fan-Rong Zhao
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Guo-Pin Pan
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Chong Zhang
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Ping Song
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Peng Li
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Jian Xu
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Shuangxi Wang
- a College of Pharmacy, Xinxiang Medical University , Xinxiang , China
| | - Ya-Ling Yin
- c School of Basic Medical Sciences, Xinxiang Medical University , Xinxiang , China
| |
Collapse
|
22
|
Fleenor BS, Ouyang A, Olver TD, Hiemstra JA, Cobb MS, Minervini G, Emter CA. Saxagliptin Prevents Increased Coronary Vascular Stiffness in Aortic-Banded Mini Swine. Hypertension 2018; 72:466-475. [PMID: 29891647 DOI: 10.1161/hypertensionaha.118.10993] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 02/25/2018] [Accepted: 05/14/2018] [Indexed: 01/09/2023]
Abstract
Increased peripheral conduit artery stiffness has been shown in patients with heart failure (HF) with preserved ejection fraction. However, it is unknown whether this phenomenon extends to the coronary vasculature. HF with preserved ejection fraction may be driven, in part, by coronary inflammation, and inhibition of the enzyme DPP-4 (dipeptidyl-peptidase 4) reduces inflammation and oxidative stress. The purpose of this study was to determine the effect of saxagliptin-a DPP-4 inhibitor-on coronary stiffness in aortic-banded mini swine. We hypothesized saxagliptin would prevent increased coronary artery stiffness in a translational swine model with cardiac features of HF with preserved ejection fraction by inhibiting perivascular adipose tissue inflammation. Yucatan mini swine were divided into 3 groups: control, aortic-banded untreated HF, and aortic-banded saxagliptin-treated HF. Ex vivo mechanical testing was performed on the left circumflex and right coronary arteries, and advanced glycation end product, NF-κB (nuclear factor-κB), and nitrotyrosine levels were measured. An increase in the coronary elastic modulus of HF animals was associated with increased vascular advanced glycation end products, NF-κB, and nitrotyrosine levels compared with control and prevented by saxagliptin treatment. Aortas from healthy mice were treated with media from swine perivascular adipose tissue culture to assess its role on vascular stiffening. Conditioned media from HF and saxagliptin-treated HF animals increased mouse aortic stiffness; however, only perivascular adipose tissue from the HF group showed increased advanced glycation end products and NF-κB levels. In conclusion, our data show increased coronary conduit vascular stiffness was prevented by saxagliptin and associated with decreased advanced glycation end products, NF-κB, and nitrotyrosine levels in a swine model with potential relevance to HF with preserved ejection fraction.
Collapse
Affiliation(s)
- Bradley S Fleenor
- From the Human Performance Laboratory, School of Kinesiology, Ball State University, Muncie, IN (B.S.F.)
| | - An Ouyang
- Department of Kinesiology and Health Promotion, University of Kentucky, Lexington (A.O.)
| | - T Dylan Olver
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| | - Jessica A Hiemstra
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| | - Melissa S Cobb
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| | | | - Craig A Emter
- Department of Biomedical Science, University of Missouri, Columbia (T.D.O., J.A.H., M.S.C., C.A.E.)
| |
Collapse
|
23
|
Battson ML, Lee DM, Jarrell DK, Hou S, Ecton KE, Weir TL, Gentile CL. Suppression of gut dysbiosis reverses Western diet-induced vascular dysfunction. Am J Physiol Endocrinol Metab 2018; 314:E468-E477. [PMID: 29351482 PMCID: PMC6048388 DOI: 10.1152/ajpendo.00187.2017] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Vascular dysfunction represents a critical preclinical step in the development of cardiovascular disease. We examined the role of the gut microbiota in the development of obesity-related vascular dysfunction. Male C57BL/6J mice were fed either a standard diet (SD) ( n = 12) or Western diet (WD) ( n = 24) for 5 mo, after which time WD mice were randomized to receive either unsupplemented drinking water or water containing a broad-spectrum antibiotic cocktail (WD + Abx) ( n = 12/group) for 2 mo. Seven months of WD caused gut dysbiosis, increased arterial stiffness (SD 412.0 ± 6.0 vs. WD 458.3 ± 9.0 cm/s, P < 0.05) and endothelial dysfunction (28% decrease in max dilation, P < 0.05), and reduced l-NAME-inhibited dilation. Vascular dysfunction was accompanied by significant increases in circulating LPS-binding protein (LBP) (SD 5.26 ± 0.23 vs. WD 11 ± 0.86 µg/ml, P < 0.05) and interleukin-6 (IL-6) (SD 3.27 ± 0.25 vs. WD 7.09 ± 1.07 pg/ml, P < 0.05); aortic expression of phosphorylated nuclear factor-κB (p-NF-κB) ( P < 0.05); and perivascular adipose expression of NADPH oxidase subunit p67phox ( P < 0.05). Impairments in vascular function correlated with reductions in Bifidobacterium spp. Antibiotic treatment successfully abrogated the gut microbiota and reversed WD-induced arterial stiffness and endothelial dysfunction. These improvements were accompanied by significant reductions in LBP, IL-6, p-NF-κB, and advanced glycation end products (AGEs), and were independent from changes in body weight and glucose tolerance. These results indicate that gut dysbiosis contributes to the development of WD-induced vascular dysfunction, and identify the gut microbiota as a novel therapeutic target for obesity-related vascular abnormalities.
Collapse
Affiliation(s)
- Micah L Battson
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| | - Dustin M Lee
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| | - Dillon K Jarrell
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| | - Shuofei Hou
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| | - Kayl E Ecton
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| | - Tiffany L Weir
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| | - Christopher L Gentile
- Department of Food Science and Human Nutrition, Colorado State University , Fort Collins, Colorado
| |
Collapse
|