1
|
Shatarat AT, Shurrab AM, Al-Lahham HM, Tarboush NA, Badran DH, Salameh MA, Badran R. Irisin Modulates Perivascular Adipose Tissue Structure In Rat Thoracic Aorta. Morphologie 2025; 109:100955. [PMID: 39985838 DOI: 10.1016/j.morpho.2025.100955] [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: 12/12/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Irisin is a newly identified hormone secreted mainly by skeletal muscles. It has different effects on the cardiovascular system and blood vessels. The present study investigated the possible effects of irisin on the microscopic structure of the perivascular adipose tissue in the rat thoracic aorta. MATERIALS AND METHODS Twenty rats were randomly divided into two groups: a non-injected control group (n=4) and four injected experimental groups (each n=4). The experimental rats were injected intraperitoneally with one of the following concentrations of irisin; 250, 500, 1000, and 2000ng/mL, twice a week for 4 weeks. After that, all experimental rats' descending thoracic aortas were removed, and imaging was performed. ANOVA and Bonferroni's Multiple Comparison Test were used to achieve statistical comparisons. RESULTS A trend of a dose-dependent increase in the number of brown adipocytes in all irisin-injected groups reached statistical significance at a dose of 2000ng/mL, compared to that of the control group (from 7.9±1 control to 67±6.1 in 2000ng/mL of irisin). A dose-dependent decrease in the number of white adipocytes compared to that of the control group (from 40±4.8 control to 3±0.9 in 2000ng/mL of irisin). CONCLUSION The present study has for the first time demonstrated that irisin has significantly increased the number of brown adipocytes and decreased the number of white adipocytes in the perivascular adipose tissue in rat thoracic aorta.
Collapse
MESH Headings
- Animals
- Aorta, Thoracic/drug effects
- Aorta, Thoracic/anatomy & histology
- Aorta, Thoracic/cytology
- Fibronectins/pharmacology
- Fibronectins/administration & dosage
- Rats
- Male
- Adipose Tissue/drug effects
- Dose-Response Relationship, Drug
- Adipose Tissue, Brown/drug effects
- Adipose Tissue, Brown/cytology
- Adipose Tissue, White/drug effects
- Adipose Tissue, White/cytology
- Rats, Wistar
- Random Allocation
- Rats, Sprague-Dawley
Collapse
Affiliation(s)
- A T Shatarat
- Aqaba Medical Sciences University, Aqaba, Jordan
| | - A M Shurrab
- Al-Balqa Applied University, Al-Salt, Jordan.
| | | | | | - D H Badran
- Ibn Sina University for Medical Sciences, Amman, Jordan
| | - M A Salameh
- Al-Balqa Applied University, Al-Salt, Jordan
| | - R Badran
- Philadelphia University, Amman, Jordan
| |
Collapse
|
2
|
Terrian L, Thompson JM, Bowman DE, Panda V, Contreras GA, Rockwell C, Sather L, Fink GD, Lauver DA, Nault R, Watts SW, Bhattacharya S. Single-nucleus analysis of thoracic perivascular adipose tissue reveals critical changes in cell composition, communication, and gene regulatory networks induced by a high fat hypertensive diet. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.13.636878. [PMID: 39990347 PMCID: PMC11844537 DOI: 10.1101/2025.02.13.636878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, with hypertension being its primary causal factor. Most blood vessels are surrounded by perivascular adipose tissue (PVAT), which regulates blood vessel tone through the secretion of vasoactive factors. PVAT is recognized as a key mediator of vascular function and dysfunction in CVD, although the underlying mechanisms remain poorly understood. To investigate PVAT's mechanistic role in hypertension, we performed single nucleus RNA-Sequencing analysis of thoracic aortic PVAT from Dahl SS rats fed a high-fat, hypertensive diet. Computational analysis revealed extensive diet-induced changes in cell-type composition, cell-type specific gene expression, cell-cell communication pathways, and intracellular gene regulatory networks within PVAT. Furthermore, we identified key transcription factors mediating these networks and demonstrated through virtual knock-out experiments that these factors could serve as potential therapeutic targets for preventing or reversing PVAT's hypertensive state.
Collapse
Affiliation(s)
- Leah Terrian
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
- Denotes individuals contributed equally as first authors to this work
| | - 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
| | - Derek E. Bowman
- 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
- College of Osteopathic Medicine, Michigan State University, East Lansing, MI, USA
| | - Vishal Panda
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - 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
| | - Lisa Sather
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Gregory D. Fink
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - D. Adam Lauver
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
| | - Rance Nault
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI, USA
| | - Stephanie W. Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, USA
- Denotes lead investigators/funding
| | - 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
- Denotes lead investigators/funding
| |
Collapse
|
3
|
Wabel EA, Krieger-Burke T, Watts SW. Vascular chemerin from PVAT contributes to norepinephrine and serotonin-induced vasoconstriction and vascular stiffness in a sex-dependent manner. Am J Physiol Heart Circ Physiol 2024; 327:H1577-H1589. [PMID: 39453435 DOI: 10.1152/ajpheart.00475.2024] [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: 07/16/2024] [Revised: 10/01/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
The adipokine chemerin supports normal blood pressure and contributes to adiposity-associated hypertension, evidenced by falls in mean arterial pressure in Dahl SS rats given an antisense oligonucleotide against chemerin. In humans, circulating chemerin is positively associated with hypertension and aortic stiffness. Mechanisms of chemerin's influence on vascular health and disease remain unknown. We identified chemerin production in the vasculature-the blood vessel and its perivascular adipose tissue (PVAT). Here, using RNAScope, qPCR, isometric contractility, high-frequency ultrasound imaging, and Western blot in the Dahl SS rat, we test the hypothesis that endogenous chemerin amplifies agonist-induced vasoconstriction through the chemerin1 receptor and that chemerin drives aortic stiffness in the thoracic aorta. CMKLR1 (chemerin1) expression was higher in the media, and Rarres2 (chemerin) expression was higher in the PVAT. Chemerin1 receptor antagonism via selective inhibitor CCX832 reduced maximal contraction to norepinephrine (NE) and serotonin (5-HT), but not angiotensin II, in isolated thoracic aorta (PVAT intact) from male Dahl SS rat. In females, CCX832 did not alter contraction to NE or 5-HT. Male, but not female, genetic chemerin knockout Dahl SS rats had lower aortic arch pulse wave velocity than wild types, indicating chemerin's role in aortic stiffness. Aortic PVAT from females expressed less chemerin protein than males, suggesting PVAT as the primary source of active chemerin. We show that chemerin made by the PVAT amplifies NE and 5-HT-induced contraction and potentially induces aortic stiffening in a sex-dependent manner, highlighting the potential for chemerin to be a key factor in blood pressure control and aortic stiffening.NEW & NOTEWORTHY Chemerin1 receptor inhibition reduced norepinephrine (NE) and 5-HT-induced vasoconstriction in males. Genetic chemerin knockout (KO) resulted in lower pulse wave velocity in males. Differences in chemerin abundance in aorta perivascular adipose tissue (APVAT) may explain sex-dependent role of chemerin.
Collapse
Affiliation(s)
- Emma A Wabel
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Teresa Krieger-Burke
- 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
| |
Collapse
|
4
|
Jenkins SW, Grunz EA, Ramos KR, Boerman EM. Perivascular Adipose Tissue Becomes Pro-Contractile and Remodels in an IL10 -/- Colitis Model of Inflammatory Bowel Disease. Int J Mol Sci 2024; 25:10726. [PMID: 39409054 PMCID: PMC11476586 DOI: 10.3390/ijms251910726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/18/2024] [Accepted: 09/29/2024] [Indexed: 10/20/2024] Open
Abstract
Inflammatory Bowel Diseases (IBDs) are associated with aberrant immune function, widespread inflammation, and altered intestinal blood flow. Perivascular adipose tissue (PVAT) surrounding the mesenteric vasculature can modulate vascular function and control the local immune cell population, but its structure and function have never been investigated in IBD. We used an IL10-/- mouse model of colitis that shares features with human IBD to test the hypothesis that IBD is associated with (1) impaired ability of PVAT to dilate mesenteric arteries and (2) changes in PVAT resident adipocyte and immune cell populations. Pressure myography and electrical field stimulation of isolated mesenteric arteries show that PVAT not only loses its anti-contractile effect but becomes pro-contractile in IBD. Quantitative immunohistochemistry and confocal imaging studies found significant adipocyte hyperplasia and increased PVAT leukocytes, particularly macrophages, in IBD. PCR arrays suggest that these changes occur alongside the altered cytokine and chemokine gene expression associated with altered NF-κB signaling. Collectively, these results show that the accumulation of macrophages in PVAT during IBD pathogenesis may lead to local inflammation, which ultimately contributes to increased arterial constriction and decreased intestinal blood flow with IBD.
Collapse
Affiliation(s)
| | | | | | - Erika M. Boerman
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|
5
|
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: 2] [Impact Index Per Article: 2.0] [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
|
6
|
Mikami T, Dashwood MR, Kawaharada N, Furuhashi M. An Obligatory Role of Perivascular Adipose Tissue in Improved Saphenous Vein Graft Patency in Coronary Artery Bypass Grafting. Circ J 2024; 88:845-852. [PMID: 37914280 DOI: 10.1253/circj.cj-23-0581] [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] [Indexed: 11/03/2023]
Abstract
The gold standard graft for coronary artery bypass grafting (CABG) is the internal thoracic artery (ITA), and the second recommendation is the radial artery. However, complete revascularization with arterial grafts alone is often difficult, and the saphenous vein (SV) is the most commonly used autologous graft for CABG, because it is easier to use without restriction for the length of the graft. On the other hand, the patency of SV grafts (SVGs) is poor compared with that of arterial grafts. The SVG is conventionally harvested as a distended conduit with surrounding tissue removed, a procedure that may cause vascular damage. A no-touch technique of SVG harvesting has been reported to result in improved long-term patency in CABG comparable to that when using the ITA for grafting. Possible reasons for the excellent long-term patency of no-touch SVGs are the physical support provided by preserved surrounding perivascular adipose tissue, preservation of the vascular wall structure including the vasa vasorum, and production of adipocyte-derived factors. In this review, we discuss recent strategies aimed at improving the performance of SVGs, including no-touch harvesting, minimally invasive harvesting and mechanical support using external stents.
Collapse
Affiliation(s)
- Takuma Mikami
- Department of Cardiovascular Surgery, Sapporo Medical University
| | - Michael R Dashwood
- Surgical and Interventional Sciences, Royal Free Hospital Campus, University College London Medical School
| | | | - Masato Furuhashi
- Department of Cardiovascular, Renal and Metabolic Medicine, Sapporo Medical University
| |
Collapse
|
7
|
Wang A, Dong S, Liu B, Liu D, Zou M, Han Y, Yang L, Wang Y. The role of RUNX1/NF-κB in regulating PVAT inflammation in aortic dissection. Sci Rep 2024; 14:9960. [PMID: 38693222 PMCID: PMC11063189 DOI: 10.1038/s41598-024-60737-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 04/26/2024] [Indexed: 05/03/2024] Open
Abstract
The pathogenesis of aortic dissection (AD), an aortic disease associated with high mortality, involves significant vascular inflammatory infiltration. However, the precise relationship between perivascular adipose tissue (PVAT) and aortic dissection remains incompletely understood. The objective of this study is to investigate the role of PVAT inflammation in the pathogenesis of aortic dissection and identify novel therapeutic targets for this disease. The mouse model of aortic dissection was established in this study through intraperitoneal injection of Ang II and administration of BAPN in drinking water. Additionally, control groups were established at different time points including the 2-week group, 3-week group, and 4-week group. qPCR and immunohistochemistry techniques were employed to detect the expression of inflammatory markers and RUNX1 in PVAT surrounding the thoracic aorta in mice. Additionally, an aortic dissection model was established using RUNX1 knockout mice, and the aforementioned indicators were assessed. The 3T3-L1 cells were induced to differentiate into mature adipocytes in vitro, followed by lentivirus transfection for the knockdown or overexpression of RUNX1. The study aimed to investigate the potential cell-to-cell interactions by co-culturing 3T3-L1 cells with A7r5 or RAW264.7 cells. Subsequently, human aortic PVAT samples were obtained through clinical surgery and the aforementioned indicators were detected. In comparison to the control group, the aortic dissection model group exhibited decreased expression of MMP-2 and NF-κB in PVAT, while TNF-α and RUNX1 expression increased. Suppression of RUNX1 expression resulted in increased MMP-2 and NF-κB expression in PVAT, along with decreased TNF-α expression. Overexpression of RUNX1 upregulated the expression levels of NF-Κb, MMP-2, and TNF-α in adipocytes, whereas knockdown of RUNX1 exerted an opposite effect. Macrophages co-cultured with adipocytes overexpressing RUNX1 exhibited enhanced CD86 expression, while vascular smooth muscle cells co-cultured with these adipocytes showed reduced α-SMA expression. In human samples, there was an increase in both RUNX1 and MMP-2 expression levels, accompanied by a decrease in TNF-α and NF-Κb expression. The presence of aortic dissection is accompanied by evident inflammatory alterations in the PVAT, and this phenomenon appears to be associated with the involvement of RUNX1. It is plausible that the regulation of PVAT's inflammatory changes by RUNX1/NF-κB signaling pathway plays a role in the pathogenesis of aortic dissection.
Collapse
Affiliation(s)
- Ao Wang
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Shengjun Dong
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Baohui Liu
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Dianxiao Liu
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Mingrui Zou
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Yuexin Han
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China
| | - Lijuan Yang
- Department of Medical Research Center, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China.
| | - Yujiu Wang
- Department of Cardiovascular Surgery, Binzhou Medical University Hospital, Binzhou, 256600, Shandong Province, China.
| |
Collapse
|
8
|
de Oliveira PB, Zochio GP, Caetano ESP, da Silva MLS, Dias-Junior CA. Vasodilator Responses of Perivascular Adipose Tissue-Derived Hydrogen Sulfide Stimulated with L-Cysteine in Pregnancy Hypertension-Induced Endothelial Dysfunction in Rats. Antioxidants (Basel) 2023; 12:1919. [PMID: 38001772 PMCID: PMC10669374 DOI: 10.3390/antiox12111919] [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: 09/25/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
Endothelium-derived nitric oxide (NO)-induced vasodilation is impaired in pregnancy hypertension. However, the role of perivascular adipose tissue (PVAT)-derived hydrogen sulfide (H2S), as an alternative for counteracting vascular dysfunction, is incompletely clear in hypertensive disorders of pregnancy. Therefore, PVAT-derived H2S-induced vasodilation was investigated in pregnancy hypertension-induced endothelial dysfunction. Non-pregnant (Non-Preg) and pregnant (Preg) rats were submitted (or not) to the deoxycorticosterone (DOCA)-salt protocol and assigned as follows (n = 10/group): Non-Preg, Non-Preg+DOCA, Preg, and Preg+DOCA groups. Systolic blood pressure (SBP), angiogenesis-related factors, determinant levels of H2S (PbS), NO (NOx), and oxidative stress (MDA) were assessed. Vascular changes were recorded in thoracic aortas with PVAT and endothelium (intact and removed layers). Vasorelaxation responses to the substrate (L-cysteine) for the H2S-producing enzyme cystathionine-γ-lyase (CSE) were examined in the absence and presence of CSE-inhibitor DL-propargylglycine (PAG) in thoracic aorta rings pre-incubated with cofactor for CSE (pyridoxal-5 phosphate: PLP) and pre-contracted with phenylephrine. Hypertension was only found in the Preg+DOCA group. Preg+DOCA rats showed angiogenic imbalances and increased levels of MDA. PbS, but not NOx, showed increased levels in the Preg+DOCA group. Pre-incubation with PLP and L-cysteine elevated determinants of H2S in PVAT and placentas of Preg-DOCA rats, whereas no changes were found in the aortas without PVAT. Aortas of Preg-DOCA rats showed that PVAT-derived H2S-dependent vasodilation was greater compared to endothelium-derived H2S, whereas PAG blocked these responses. PVAT-derived H2S endogenously stimulated with the amino acid L-cysteine may be an alternative to induce vasorelaxation in endothelial dysfunction related to pregnancy hypertension.
Collapse
Affiliation(s)
- Priscilla Bianca de Oliveira
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (P.B.d.O.); (G.P.Z.); (E.S.P.C.); (M.L.S.d.S.)
- Laboratory of Pharmacology, Marília Medical School (FAMEMA), Marília 17519-030, SP, Brazil
| | - Gabriela Palma Zochio
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (P.B.d.O.); (G.P.Z.); (E.S.P.C.); (M.L.S.d.S.)
| | - Edileia Souza Paula Caetano
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (P.B.d.O.); (G.P.Z.); (E.S.P.C.); (M.L.S.d.S.)
| | - Maria Luiza Santos da Silva
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (P.B.d.O.); (G.P.Z.); (E.S.P.C.); (M.L.S.d.S.)
| | - Carlos Alan Dias-Junior
- Department of Biophysics and Pharmacology, Institute of Biosciences, São Paulo State University (UNESP), Botucatu 18618-689, SP, Brazil; (P.B.d.O.); (G.P.Z.); (E.S.P.C.); (M.L.S.d.S.)
| |
Collapse
|
9
|
Cai M, Zhao D, Han X, Han S, Zhang W, Zang Z, Gai C, Rong R, Gao T. The role of perivascular adipose tissue-secreted adipocytokines in cardiovascular disease. Front Immunol 2023; 14:1271051. [PMID: 37822930 PMCID: PMC10562567 DOI: 10.3389/fimmu.2023.1271051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 08/28/2023] [Indexed: 10/13/2023] Open
Abstract
Perivascular adipose tissue and the vessel wall are connected through intricate bidirectional paracrine and vascular secretory signaling pathways. The secretion of inflammatory factors and oxidative products by the vessel wall in the diseased segment has the ability to influence the phenotype of perivascular adipocytes. Additionally, the secretion of adipokines by perivascular adipose tissue exacerbates the inflammatory response in the diseased vessel wall. Therefore, quantitative and qualitative studies of perivascular adipose tissue are of great value in the context of vascular inflammation and may provide a reference for the assessment of cardiovascular ischemic disease.
Collapse
Affiliation(s)
- Meichao Cai
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dongsheng Zhao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Han
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Shuang Han
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Wenxin Zhang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhennan Zang
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Chenchen Gai
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Rong Rong
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tian Gao
- School of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
10
|
Ahmed A, Bibi A, Valoti M, Fusi F. Perivascular Adipose Tissue and Vascular Smooth Muscle Tone: Friends or Foes? Cells 2023; 12:cells12081196. [PMID: 37190105 DOI: 10.3390/cells12081196] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/09/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Perivascular adipose tissue (PVAT) is a specialized type of adipose tissue that surrounds most mammalian blood vessels. PVAT is a metabolically active, endocrine organ capable of regulating blood vessel tone, endothelium function, vascular smooth muscle cell growth and proliferation, and contributing critically to cardiovascular disease onset and progression. In the context of vascular tone regulation, under physiological conditions, PVAT exerts a potent anticontractile effect by releasing a plethora of vasoactive substances, including NO, H2S, H2O2, prostacyclin, palmitic acid methyl ester, angiotensin 1-7, adiponectin, leptin, and omentin. However, under certain pathophysiological conditions, PVAT exerts pro-contractile effects by decreasing the production of anticontractile and increasing that of pro-contractile factors, including superoxide anion, angiotensin II, catecholamines, prostaglandins, chemerin, resistin, and visfatin. The present review discusses the regulatory effect of PVAT on vascular tone and the factors involved. In this scenario, dissecting the precise role of PVAT is a prerequisite to the development of PVAT-targeted therapies.
Collapse
Affiliation(s)
- Amer Ahmed
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Aasia Bibi
- Nanotechnology Institute, CNR-NANOTEC, Via Monteroni, 73100 Lecce, Italy
| | - Massimo Valoti
- Department of Life Sciences, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| | - Fabio Fusi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy
| |
Collapse
|
11
|
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: 26] [Impact Index Per Article: 13.0] [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
|
12
|
Zhang QH, Li Y, Xie LH, Ren X, Zhao Y, Wang N, Chen LH, Ju Y, Liu YJ, Xie LZ, Lin LJ, Liu AL. Association Between Perivascular Adipose Tissue Density and Atherosclerosis in the Descending Thoracic Aorta. Angiology 2023; 74:216-226. [PMID: 35500088 DOI: 10.1177/00033197221098894] [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: 02/01/2023]
Abstract
Radiodensity measured by computed tomography (CT) in Hounsfield Units (HU) is emerging as a clinical tool for detecting perivascular adipose tissue (PVAT) inflammation. In the present study, we hypothesized that PVAT radiodensity might predict the risk of descending thoracic aorta atherosclerosis. A total of 73 subjects who underwent CT angiography to investigate aortic disease were retrospectively analyzed. PVAT radiodensity, aortic complex plaque (ACP), mean plaque-burden score (MPBS), and plaque density were measured, and the association between them was analyzed. Perivascular adipose tissue radiodensity (HU) in patients with different aortic plaques grades (grade 1, 2, 3, and 4) were -93.71 ± 2.50, -93.63 ± 3.93, -90.24 ± 4.49, and -89.90 ± 5.18, respectively, and the difference was significant (P = .010). In the regression analysis, PVAT radiodensity was an independent predictor of ACP, with an OR of 1.263. In the linear analysis, PVAT radiodensity was an independent predictor of MPBS, with a β-coefficient of .073. In the univariate analysis, only the PVAT radiodensity was significantly associated with plaque density, with a β-coefficient of -1.666. In conclusion, PVAT density was independently related to descending thoracic aorta atherosclerosis.
Collapse
Affiliation(s)
- Qin-He Zhang
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ye Li
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Lu-Han Xie
- Department of Pathology and Forensics, 36674Dalian Medical University, Dalian, China
| | - Xue Ren
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ying Zhao
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Nan Wang
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li-Hua Chen
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ye Ju
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yi-Jun Liu
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Li-Zhi Xie
- MR Research, 33112GE Healthcare, Beijing, China
| | - Liang-Jie Lin
- 244810Clinical and Technical Support, Philips Healthcare, Beijing, China
| | - Ai-Lian Liu
- Department of Radiology, 74710the First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
13
|
Lázaro-Suárez ML, Domínguez de la Mora I, Rodríguez-Aguilar JC, Fortis-Barrera Á, Blancas-Flores G, Gómez-Zamudio JH, Alarcon-Villaseñor EF, Román-Ramos R, Alarcon-Aguilar FJ. Role of Perivascular Adipose Tissue in Aorta Reactivity from Obese and Hyperglycemic CD-1 Mice: New Insights into Perivascular Adipose Tissue. Metab Syndr Relat Disord 2023; 21:101-108. [PMID: 36399542 DOI: 10.1089/met.2022.0050] [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: 11/19/2022] Open
Abstract
Background: Perivascular adipose tissue (PVAT) plays an essential role in cardiovascular homeostasis. However, during obesity and diabetes, its role in vascular tone regulation is unclear. This study aimed to evaluate the function of the PVAT on aorta reactivity in the lean and cafeteria (CAF) diet-induced obese-hyperglycemic mice model. Methods: Aorta reactivity to phenylephrine, KCl, and acetylcholine was analyzed in lean (n = 6) and obese mice (n = 6). Also, nitric oxide (NO-) and cyclooxygenase participation, in the presence (n = 6) and absence (n = 6) of PVAT, were examined in the aortas. Results: After a CAF diet for 19 weeks, obese mice showed increased body weight, glucose intolerance, and hypercholesterolemia concerning lean mice. Vascular reactivity to phenylephrine was reduced significantly in the aorta of obese mice. In contrast, the contraction produced by KCl (80 mM) was increased in the aorta of obese mice independent of PVAT. Acetylcholine-induced vasorelaxation diminished in the aortas of obese mice in the presence of PVAT. Nonselective inhibition of cyclooxygenases likely shows that PVAT and endothelium release vasorelaxant prostanoids. Conclusions: The results suggest that PVAT modulates aorta reactivity by releasing NO-, decreasing the α1-adrenergic response to phenylephrine, and probably releasing vasorelaxant prostanoids. The data suggest that PVAT regulates the vascular smooth muscle and endothelial function in a CAF diet-induced obese-hyperglycemic mice model.
Collapse
Affiliation(s)
- Martha L Lázaro-Suárez
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Israel Domínguez de la Mora
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Juan Carlos Rodríguez-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Ángeles Fortis-Barrera
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Gerardo Blancas-Flores
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Jaime H Gómez-Zamudio
- Unidad de Investigación Médica en Bioquímica, Hospital de Especialidades, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Ciudad de Mexico, Mexico
| | | | - Rubén Román-Ramos
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| | - Francisco Javier Alarcon-Aguilar
- Laboratorio de Farmacología, Departamento de Ciencias de la Salud, DCBS, Universidad Autónoma Metropolitana-Iztapalapa, Ciudad de México, México
| |
Collapse
|
14
|
Márquez-Sánchez AC, Koltsova EK. Immune and inflammatory mechanisms of abdominal aortic aneurysm. Front Immunol 2022; 13:989933. [PMID: 36275758 PMCID: PMC9583679 DOI: 10.3389/fimmu.2022.989933] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/23/2022] [Indexed: 11/13/2022] Open
Abstract
Abdominal aortic aneurysm (AAA) is a life-threatening cardiovascular disease. Immune-mediated infiltration and a destruction of the aortic wall during AAA development plays significant role in the pathogenesis of this disease. While various immune cells had been found in AAA, the mechanisms of their activation and function are still far from being understood. A better understanding of mechanisms regulating the development of aberrant immune cell activation in AAA is essential for the development of novel preventive and therapeutic approaches. In this review we summarize current knowledge about the role of immune cells in AAA and discuss how pathogenic immune cell activation is regulated in this disease.
Collapse
|
15
|
Xie X, Wang S, Rao J, Xue J, Lin K, Lin N, Li K, Wu S, Liang W, Guo Y. Comprehensive Analysis of Differentially Expressed lncRNAs in the Perivascular Adipose Tissue of Patients with Coronary Heart Disease. Rev Cardiovasc Med 2022; 23:341. [PMID: 39077137 PMCID: PMC11267359 DOI: 10.31083/j.rcm2310341] [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: 04/04/2022] [Revised: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 07/31/2024] Open
Abstract
Background Coronary heart disease is a highly prevalent inflammatory disease caused by coronary atherosclerosis. Numerous studies have revealed that perivascular adipose tissue is closely associated with atherosclerosis. Here, we conducted a comprehensive analysis of long non-coding RNAs and mRNAs differentially expressed in perivascular adipose tissue in patients with coronary heart disease. Methods We conducted Gene Ontology term and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analyses of the differentially expressed genes. Furthermore, single sample gene set enrichment analysis, immune infiltration analysis, and co-expression analysis of differentially expressed long non-coding RNAs and immune gene sets were performed. Finally, the starBase and miRTarBase databases were used to construct a competing endogenous RNA network. Results The results show that aortic perivascular adipose tissue has higher inflammation and immune infiltration levels in patients with coronary heart disease. Dysregulated long non-coding RNAs may be related to immunity, inflammation, and hypoxia. Conclusions The findings of this study provide new insights into atherosclerosis and coronary heart disease.
Collapse
Affiliation(s)
- Xianwei Xie
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
| | - Sunying Wang
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
| | - Jingyi Rao
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
| | - Jing Xue
- Beijing Tiantan Hospital, China National Clinical Research Center of Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, 100070 Beijing, China
| | - Kaiyang Lin
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Clinical Medical Research Center for cardiovascular diseases, 350000 Fuzhou, Fujian, China
- Fujian Heart Failure Center Alliance, 350000 Fuzhou, Fujian, China
| | - Na Lin
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Clinical Medical Research Center for cardiovascular diseases, 350000 Fuzhou, Fujian, China
| | - Ke Li
- Beijing Tiantan Hospital, China National Clinical Research Center of Neurological Diseases, Advanced Innovation Center for Human Brain Protection, The Capital Medical University, 100070 Beijing, China
| | - Shilun Wu
- Department of Hepatobiliary Surgery, Beijing Chaoyang Hospital Affiliated to Capital Medical University, 100043 Beijing, China
| | - Wenjia Liang
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
| | - Yansong Guo
- Department of Cardiology, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, 350013 Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Cardiovascular Disease, Fujian Provincial Center for Geriatrics, Fujian Clinical Medical Research Center for cardiovascular diseases, 350000 Fuzhou, Fujian, China
- Fujian Heart Failure Center Alliance, 350000 Fuzhou, Fujian, China
| |
Collapse
|
16
|
Mitidieri E, Turnaturi C, Vanacore D, Sorrentino R, d'Emmanuele di Villa Bianca R. The Role of Perivascular Adipose Tissue-Derived Hydrogen Sulfide in the Control of Vascular Homeostasis. Antioxid Redox Signal 2022; 37:84-97. [PMID: 35442088 DOI: 10.1089/ars.2021.0147] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Significance: Emerging evidence suggests that perivascular adipose tissue (PVAT) has a relevant role in the control of vascular tone in physiology and pathology. Healthy PVAT has anticontractile, anti-inflammatory, and antioxidative actions. Accumulating data from both human and experimental animal models indicate that PVAT dysfunction is conceivably coupled to cardiovascular diseases, and it is associated with vascular inflammation, oxidative stress, and arterial remodeling. Therefore, "healthy" PVAT may constitute a novel therapeutic target for the prevention and treatment of cardiovascular diseases. Recent Advances: Hydrogen sulfide (H2S) has been recognized as a vascular anti-contractile factor released from PVAT. The enzymes deputed to H2S biosynthesis are variously expressed in PVAT and strictly dependent on the vascular bed and species. Metabolic and cardiovascular diseases can alter the morphological and secretory characteristics of PVAT, influencing also the H2S signaling. Here, we discuss the role of PVAT-derived H2S in healthy conditions and its relevance in alterations occurring in vascular disorders. Critical Issues: We discuss how a better understanding may help in the prevention of vascular dysfunction related to alteration in PVAT-released H2S as well as the importance of the interplay between PVAT and H2S. Future Directions: We propose future directions to evaluate the contribution of each enzyme involved in H2S biosynthesis and their alteration/switch occurring in vascular disorders and the remaining challenges in investigating the role of H2S. Antioxid. Redox Signal. 37, 84-97.
Collapse
Affiliation(s)
- Emma Mitidieri
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Carlotta Turnaturi
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Domenico Vanacore
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Raffaella Sorrentino
- Department of Molecular Medicine and Medical Biotechnology, School of Medicine, University of Naples Federico II, Naples, Italy
| | | |
Collapse
|
17
|
Kagota S, Futokoro R, McGuire JJ, Maruyama-Fumoto K, Shinozuka K. Modulation of Vasomotor Function by Perivascular Adipose Tissue of Renal Artery Depends on Severity of Arterial Dysfunction to Nitric Oxide and Severity of Metabolic Parameters. Biomolecules 2022; 12:biom12070870. [PMID: 35883426 PMCID: PMC9312868 DOI: 10.3390/biom12070870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/17/2022] [Accepted: 06/21/2022] [Indexed: 02/04/2023] Open
Abstract
Perivascular adipose tissue (PVAT) enhances vascular relaxation of mesenteric arteries in SHRSP.Z-Leprfa/IzmDmcr rats (SPZF), a metabolic syndrome model. We investigated and compared the effects of PVAT on the renal artery in SPZF with those on SHR/NDmcr-cp rats (CP). Renal arteries with and without PVAT were isolated from 23-week-old SPZF and CP. The effects of PVAT on acetylcholine- and nitroprusside-induced relaxation were examined using bioassays with phenylephrine-contracted arterial rings. Acetylcholine-induced relaxations without PVAT in SPZF and CP were 0.7- and 0.5-times lower in females than in males, respectively. In the presence of PVAT, acetylcholine-induced relaxations increased 1.4- and 2-times in male and female CP, respectively, but did not differ in SPZF. Nitroprusside-induced relaxation with and without PVAT was 0.7-times lower in female than in male SPZF but did not differ in CP. Angiotensin-II type-1 receptor (AT1R)/AT1R-associated protein mRNA ratios were lower in CP than in the SPZF and negatively correlated with the difference in arterial relaxation with and without PVAT. The effects of renal artery PVAT differed between the SPZF and CP groups. Higher levels of enhanced AT1R activity in SPZF PVAT may drive these differences by impairing the vascular smooth muscle responses to nitric oxide.
Collapse
Affiliation(s)
- Satomi Kagota
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya 663 8179, Japan; (R.F.); (K.M.-F.); (K.S.)
- Institute for Bioscience, Mukogawa Women’s University, Nishinomiya 663 8179, Japan
- Correspondence: ; Tel.: +81-798-45-9944
| | - Risa Futokoro
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya 663 8179, Japan; (R.F.); (K.M.-F.); (K.S.)
| | - John J. McGuire
- Departments of Medical Biophysics, Physiology & Pharmacology, Schulich School of Medicine & Dentistry, Western University, London, ON N6A 5C1, Canada;
| | - Kana Maruyama-Fumoto
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya 663 8179, Japan; (R.F.); (K.M.-F.); (K.S.)
| | - Kazumasa Shinozuka
- Department of Pharmacology, School of Pharmacy and Pharmaceutical Sciences, Mukogawa Women’s University, Nishinomiya 663 8179, Japan; (R.F.); (K.M.-F.); (K.S.)
| |
Collapse
|
18
|
Kagota S, Futokoro R, Maruyama-Fumoto K, McGuire JJ, Shinozuka K. Perivascular Adipose Tissue Compensation for Endothelial Dysfunction in the Superior Mesenteric Artery of Female SHRSP.Z-<i>Lepr</i><sup><i>fa</i></sup>/IzmDmcr Rats. J Vasc Res 2022; 59:209-220. [DOI: 10.1159/000524187] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/14/2022] [Indexed: 12/17/2022] Open
Abstract
Regulation of arterial tone by perivascular adipose tissue (PVAT) differs between sexes. In male SHRSP.Z-<i>Lepr</i><sup><i>fa</i></sup>/IzmDmcr rats (SHRSP.ZF), PVAT exerts a compensatory relaxation effect for the loss of endothelium-mediated vasorelaxation, which occurs during the early stages of metabolic syndrome. However, this effect deteriorates by 23 weeks of age. Here, therefore, we compared the effects of PVAT in female and male SHRSP.ZF. Acetylcholine-induced relaxation in superior mesenteric artery without PVAT did not differ between 23-week-old females and males. However, the presence of PVAT enhanced relaxation in 23-week-old females, but not in males. The mRNA levels of angiotensin II type 1 receptor (AT1R) in PVAT did not differ between sexes, but AT1R-associated protein (ATRAP) and apelin levels were higher in females than in males. We observed a positive relationship between differences in artery relaxation with and without PVAT and ATRAP or apelin mRNA levels. In 30-week-old females, PVAT-enhanced relaxation disappeared, and mRNA levels of AT1R increased, while apelin levels decreased compared to 23-week-old females. These results demonstrated that in SHRSP.ZF, PVAT compensation for endothelium dysfunction extended to older ages in females than in males. Apelin and AT1R/ATRAP expression in PVAT may be predictors of favorable effects.
Collapse
|
19
|
Amer A, Fabio F, Valoti M. Perivascular Adipose Tissue Modulates the Effects of Flavonoids on Rat Aorta Rings: Role of Superoxide Anion and β3 Receptors. Pharmacol Res 2022; 180:106231. [DOI: 10.1016/j.phrs.2022.106231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/14/2022] [Accepted: 04/19/2022] [Indexed: 12/23/2022]
|
20
|
Tuttle T, Darios E, Watts SW, Roccabianca S. Aortic stiffness is lower when perivascular adipose tissue (PVAT) is included: a novel ex vivo mechanics study. Am J Physiol Heart Circ Physiol 2022; 322:H1003-H1013. [PMID: 35275760 DOI: 10.1152/ajpheart.00574.2021] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Perivascular adipose tissue (PVAT) is increasingly recognized as an essential layer of the functional vasculature, being responsible for producing vasoactive substances and assisting arterial stress relaxation. Here we test the hypothesis that PVAT reduces aortic stiffness. Our model was the thoracic aorta of the male Sprague Dawley rat. Uniaxial mechanical tests for three groups of tissue were performed: aorta +PVAT (+PVAT), aorta - PVAT (-PVAT), and isolated PVAT (PVAT only). The output of the mechanical test is reported in the form of a Cauchy stress-stretch curve. This work presents a novel, physiologically relevant approach to measure mechanical stiffness ex vivo in isolated PVAT. Low-stress stiffness (), high-stress stiffness (), and the stress corresponding to a stretch of 1.2 () were measured as metrics of distensibility. The low-stress stiffness was largest in the -PVAT samples and smallest in PVAT only samples. Both the high-stress stiffness and the stress at 1.2 stretch were significantly higher in -PVAT samples when compared to +PVAT samples. Taken together these results suggest that -PVAT samples are stiffer (less distensible) both at low stress (not significant) as well as at high stress (significant) when compared to +PVAT samples. These conclusions are supported by the results of the continuum mechanics material model we also used to interpret the same experimental data. Thus, tissue stiffness is significantly lower when considering PVAT as part of the aortic wall. As such, PVAT should be considered as a target for improving vascular function in diseases with elevated aortic stiffness, including hypertension.
Collapse
Affiliation(s)
- Tyler Tuttle
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| | - Emma Darios
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI, United States
| | - Sara Roccabianca
- Department of Mechanical Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
21
|
Rami AZA, Hamid AA, Anuar NNM, Aminuddin A, Ugusman A. Exploring the Relationship of Perivascular Adipose Tissue Inflammation and the Development of Vascular Pathologies. Mediators Inflamm 2022; 2022:2734321. [PMID: 35177953 PMCID: PMC8846975 DOI: 10.1155/2022/2734321] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 01/11/2022] [Accepted: 01/21/2022] [Indexed: 12/18/2022] Open
Abstract
Initially thought to only provide mechanical support for the underlying blood vessels, perivascular adipose tissue (PVAT) has now emerged as a regulator of vascular function. A healthy PVAT exerts anticontractile and anti-inflammatory actions on the underlying vasculature via the release of adipocytokines such as adiponectin, nitric oxide, and omentin. However, dysfunctional PVAT produces more proinflammatory adipocytokines such as leptin, resistin, interleukin- (IL-) 6, IL-1β, and tumor necrosis factor-alpha, thus inducing an inflammatory response that contributes to the pathogenesis of vascular diseases. In this review, current knowledge on the role of PVAT inflammation in the development of vascular pathologies such as atherosclerosis and hypertension was discussed.
Collapse
Affiliation(s)
- Afifah Zahirah Abd Rami
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Adila A. Hamid
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Nur Najmi Mohamad Anuar
- Center for Toxicology & Health Risk Studies, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abd Aziz, 50300 Kuala Lumpur, Malaysia
| | - Amilia Aminuddin
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| | - Azizah Ugusman
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Cheras, 56000 Kuala Lumpur, Malaysia
| |
Collapse
|
22
|
Verdú E, Homs J, Boadas-Vaello P. Physiological Changes and Pathological Pain Associated with Sedentary Lifestyle-Induced Body Systems Fat Accumulation and Their Modulation by Physical Exercise. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:13333. [PMID: 34948944 PMCID: PMC8705491 DOI: 10.3390/ijerph182413333] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 12/02/2021] [Accepted: 12/10/2021] [Indexed: 12/11/2022]
Abstract
A sedentary lifestyle is associated with overweight/obesity, which involves excessive fat body accumulation, triggering structural and functional changes in tissues, organs, and body systems. Research shows that this fat accumulation is responsible for several comorbidities, including cardiovascular, gastrointestinal, and metabolic dysfunctions, as well as pathological pain behaviors. These health concerns are related to the crosstalk between adipose tissue and body systems, leading to pathophysiological changes to the latter. To deal with these health issues, it has been suggested that physical exercise may reverse part of these obesity-related pathologies by modulating the cross talk between the adipose tissue and body systems. In this context, this review was carried out to provide knowledge about (i) the structural and functional changes in tissues, organs, and body systems from accumulation of fat in obesity, emphasizing the crosstalk between fat and body tissues; (ii) the crosstalk between fat and body tissues triggering pain; and (iii) the effects of physical exercise on body tissues and organs in obese and non-obese subjects, and their impact on pathological pain. This information may help one to better understand this crosstalk and the factors involved, and it could be useful in designing more specific training interventions (according to the nature of the comorbidity).
Collapse
Affiliation(s)
- Enrique Verdú
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| | - Judit Homs
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
- Department of Physical Therapy, EUSES-University of Girona, 17190 Salt, Spain
| | - Pere Boadas-Vaello
- Research Group of Clinical Anatomy, Embryology and Neuroscience (NEOMA), Department of Medical Sciences, University of Girona, 17003 Girona, Spain;
| |
Collapse
|
23
|
Balakumar P, Alqahtani A, Khan NA, Alqahtani T, A T, Jagadeesh G. The physiologic and physiopathologic roles of perivascular adipose tissue and its interactions with blood vessels and the renin-angiotensin system. Pharmacol Res 2021; 173:105890. [PMID: 34536547 DOI: 10.1016/j.phrs.2021.105890] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/05/2021] [Accepted: 09/12/2021] [Indexed: 01/16/2023]
Abstract
The perivascular adipose tissue (PVAT) refers to an ectopic local deposit of connective tissue that anatomically surrounds most of the blood vessels. While it was initially known only as a structural support for vasculature, the landmark findings of Soltis and Cassis (1991), first demonstrating that PVAT reduces the contractions of norepinephrine in the isolated rat aorta, brought the potential vascular role of PVAT into the limelight. This seminal work implied the potential ability of PVAT to influence vascular responsiveness. Several vasoactive/vasocrine substances influencing vascular homeostasis were successively shown to be released from PVAT that include both adipocyte-derived relaxing and contracting factors. The PVAT is currently recognized as a metabolically active endocrine organ and is eventually considered as the 'protagonist' in vascular homeostasis. It plays prominent defending and opposing roles in vascular function, while the actual vascular influences of PVAT vary with an increase in adiposity. Recent studies have presented compelling evidence implicating the pivotal role of PVAT in the local activation of the renin-angiotensin system (RAS), which substantially impacts vascular physiology and physiopathology. Current findings have advanced our understanding of the role of PVAT in favorably or adversely modulating the vascular function through differential RAS activation. Given that adipocytes also produce major RAS components locally to influence vascular function, this review provides a scientific basis to distinctly understand the key role of PVAT in regulating the autocrine and paracrine functions of vascular RAS components and its potential as an emerging therapeutic target for mitigating cardiovascular complications.
Collapse
Affiliation(s)
- Pitchai Balakumar
- Department of Pharmacology, Pannai College of Pharmacy, Dindigul 624005, India.
| | - Ali Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Guraiger, Abha 62529, Kingdom of Saudi Arabia
| | - Noohu Abdulla Khan
- Department of Clinical Pharmacy, College of Pharmacy, King Khalid University, Guraiger, Abha 62529, Kingdom of Saudi Arabia
| | - Taha Alqahtani
- Department of Pharmacology, College of Pharmacy, King Khalid University, Guraiger, Abha 62529, Kingdom of Saudi Arabia
| | - Thangathirupathi A
- Department of Pharmacology, Pannai College of Pharmacy, Dindigul 624005, India
| | - Gowraganahalli Jagadeesh
- Division of Pharmacology & Toxicology, Office of Cardiology, Hematology, Endocrinology, and Nephrology, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
24
|
Meyer MR, Barton M. Role of Perivascular Adipose Tissue for Sex Differences in Coronary Artery Disease and Spontaneous Coronary Artery Dissection (SCAD). ENDOCRINE AND METABOLIC SCIENCE 2021. [DOI: 10.1016/j.endmts.2020.100068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
|
25
|
Bercea CI, Cottrell GS, Tamagnini F, McNeish AJ. Omega-3 polyunsaturated fatty acids and hypertension: a review of vasodilatory mechanisms of docosahexaenoic acid and eicosapentaenoic acid. Br J Pharmacol 2021; 178:860-877. [PMID: 33283269 DOI: 10.1111/bph.15336] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 02/06/2023] Open
Abstract
Hypertension is often characterised by impaired vasodilation involving dysfunction of multiple vasodilatory mechanisms. ω-3 polyunsaturated fatty acids (PUFAs), docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) can reduce blood pressure and vasodilation. In the endothelium, DHA and EPA improve function including increased NO bioavailability. However, animal studies show that DHA- and EPA-mediated vasodilation persists after endothelial removal, indicating a role for vascular smooth muscle cells (VSMCs). The vasodilatory effects of ω-3 PUFAs on VSMCs are mediated via opening of large conductance calcium-activated potassium channels (BKCa ), ATP-sensitive potassium channels (KATP ) and possibly members of the Kv 7 family of voltage-activated potassium channels, resulting in hyperpolarisation and relaxation. ω-3 PUFA actions on BKCa and voltage-gated ion channels involve electrostatic interactions that are dependent on the polyunsaturated acyl tail, cis-geometry of these double bonds and negative charge of the carboxyl headgroup. This suggests structural manipulation of ω-3 PUFA could generate novel, targeted, therapeutic leads.
Collapse
Affiliation(s)
- Cristiana-Ioana Bercea
- Reading School of Pharmacy, School of Chemistry, Food and Pharmacy, The University of Reading, Reading, UK
| | - Graeme S Cottrell
- Reading School of Pharmacy, School of Chemistry, Food and Pharmacy, The University of Reading, Reading, UK
| | - Francesco Tamagnini
- Reading School of Pharmacy, School of Chemistry, Food and Pharmacy, The University of Reading, Reading, UK
| | - Alister J McNeish
- Reading School of Pharmacy, School of Chemistry, Food and Pharmacy, The University of Reading, Reading, UK
| |
Collapse
|
26
|
Loesch A, Dashwood MR. Saphenous Vein Vasa Vasorum as a Potential Target for Perivascular Fat-Derived Factors. Braz J Cardiovasc Surg 2020; 35:964-969. [PMID: 33306322 PMCID: PMC7731844 DOI: 10.21470/1678-9741-2020-0031] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is a source of factors affecting vasomotor tone with the potential to play a role in the performance of saphenous vein (SV) bypass grafts. As these factors have been described as having constrictor or relaxant effects, they may be considered either beneficial or detrimental. The close proximity of PVAT to the adventitia provides an environment whereby adipose tissue-derived factors may affect the vasa vasorum, a microvascular network providing the vessel wall with oxygen and nutrients. Since medial ischaemia promotes aspects of graft occlusion the involvement of the PVAT/vasa vasorum axis in vein graft patency should be considered.
Collapse
Affiliation(s)
- Andrzej Loesch
- Centre for Rheumatology and Connective Tissue Diseases, University College London Medical School, Royal Free Campus, London, United Kingdom
| | - Michael Richard Dashwood
- Division of Surgery and Interventional Science, University College London Medical School, Royal Free Campus, London, United Kingdom
| |
Collapse
|
27
|
Myeloid GRK2 Regulates Obesity-Induced Endothelial Dysfunction by Modulating Inflammatory Responses in Perivascular Adipose Tissue. Antioxidants (Basel) 2020; 9:antiox9100953. [PMID: 33020373 PMCID: PMC7600489 DOI: 10.3390/antiox9100953] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 09/30/2020] [Indexed: 12/23/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is increasingly being regarded as an important endocrine organ that directly impacts vessel function, structure, and contractility in obesity-associated diseases. We uncover here a role for myeloid G protein-coupled receptor kinase 2 (GRK2) in the modulation of PVAT-dependent vasodilation responses. GRK2 expression positively correlates with myeloid- (CD68) and lymphoid-specific (CD3, CD4, and CD8) markers and with leptin in PVAT from patients with abdominal aortic aneurysms. Using mice hemizygous for GRK2 in the myeloid lineage (LysM-GRK2+/−), we found that GRK2 deficiency in myeloid cells allows animals to preserve the endothelium-dependent acetylcholine or insulin-induced relaxation, which is otherwise impaired by PVAT, in arteries of animals fed a high fat diet (HFD). Downregulation of GRK2 in myeloid cells attenuates HFD-dependent infiltration of macrophages and T lymphocytes in PVAT, as well as the induction of tumor necrosis factor-α (TNFα) and NADPH oxidase (Nox)1 expression, whereas blocking TNFα or Nox pathways by pharmacological means can rescue the impaired vasodilator responses to insulin in arteries with PVAT from HFD-fed animals. Our results suggest that myeloid GRK2 could be a potential therapeutic target in the development of endothelial dysfunction induced by PVAT in the context of obesity.
Collapse
|
28
|
Wahab A, Dey AK, Bandyopadhyay D, Katikineni V, Chopra R, Vedantam KS, Devraj M, Chowdary AK, Navarengom K, Lavie CJ, Kolpakchi A, Jneid H. Obesity, Systemic Hypertension, and Pulmonary Hypertension: A Tale of Three Diseases. Curr Probl Cardiol 2020; 46:100599. [PMID: 32560908 DOI: 10.1016/j.cpcardiol.2020.100599] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2020] [Accepted: 04/06/2020] [Indexed: 12/11/2022]
Abstract
Cardiovascular disease (CVD), especially ischemic heart disease and stroke, is the major cause of death worldwide, accounting for more than one-third of all deaths annually. Hypertension is the most prevalent and modifiable risk factor of CVD-related deaths. The same is true for obesity, which is currently being recognized as a major global epidemic. The prevalence of obesity in the United States has increased dramatically, from 13.4% in 1960 to 36.5% in 2014, with as much as 70.7% of the American adult population being overweight or obese (CDC). Epidemiological studies have shown that obesity predisposes to hypertension and CVD - with the relationship between markers of obesity and blood pressure being almost linear across different populations. In this review, we discuss systemic and pulmonary hypertension in the context of obesity.
Collapse
Affiliation(s)
- Abdul Wahab
- University of Iowa Hospitals and Clinics, Iowa City, IA
| | - Amit K Dey
- National Heart, Lung and Blood Institute, Bethesda, MD
| | | | | | | | | | | | | | | | - Carl J Lavie
- John Ochsner Heart and Vascular Institute, Ochsner Clinical School-UQ School of Medicine, New Orleans, LA
| | - Anna Kolpakchi
- Section of Cardiology, Baylor College of Medicine and the Michael E. DeBakey VAMC, Houston, TX
| | - Hani Jneid
- Section of Cardiology, Baylor College of Medicine and the Michael E. DeBakey VAMC, Houston, TX.
| |
Collapse
|
29
|
Bellner L, Lebovics NB, Rubinstein R, Buchen YD, Sinatra E, Sinatra G, Abraham NG, McClung JA, Thompson EA. Heme Oxygenase-1 Upregulation: A Novel Approach in the Treatment of Cardiovascular Disease. Antioxid Redox Signal 2020; 32:1045-1060. [PMID: 31891663 PMCID: PMC7153645 DOI: 10.1089/ars.2019.7970] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Heme oxygenase (HO) plays a pivotal role in both vascular and metabolic functions and is involved in many physiological and pathophysiological processes in vascular endothelial cells (ECs) and adipocytes. Recent Advances: From the regulation of adipogenesis in adipose tissue to the adaptive response of vascular tissue in the ECs, HO plays a critical role in the capability of the vascular system to respond and adjust to insults in homeostasis. Recent studies show that HO-1 through regulation of adipocyte and adipose tissue functions ultimately aid not only in local but also in systemic maintenance of homeostasis. Critical Issues: Recent advances have revealed the existence of a cross talk between vascular ECs and adipocytes in adipose tissue. In the pathological state of obesity, this cross talk contributes to the condition's adverse chronic effects, and we propose that specific targeting of the HO-1 gene can restore signaling pathways and improve both vascular and adipose functions. Future Directions: A complete understanding of the role of HO-1 in regulation of cardiovascular homeostasis is important to comprehend the homeostatic regulation as well as in cardiovascular disease. Efforts are required to highlight the effects and the ability to target the HO-1 gene in models of obesity with an emphasis on the role of pericardial fat on cardiovascular health.
Collapse
Affiliation(s)
- Lars Bellner
- Department of Pharmacology and New York Medical College, Valhalla, New York
| | - Nachum B Lebovics
- Department of Pharmacology and New York Medical College, Valhalla, New York
| | | | - Yosef D Buchen
- Department of Pharmacology and New York Medical College, Valhalla, New York
| | - Emilia Sinatra
- Department of Pharmacology and New York Medical College, Valhalla, New York
| | - Giuseppe Sinatra
- Department of Pharmacology and New York Medical College, Valhalla, New York
| | - Nader G Abraham
- Department of Pharmacology and New York Medical College, Valhalla, New York.,Department of Medicine, New York Medical College, Valhalla, New York
| | - John A McClung
- Department of Medicine, New York Medical College, Valhalla, New York
| | - Ellen A Thompson
- Department of Medicine, Marshall University, Joan C. Edwards School of Medicine, Huntington, West Virginia
| |
Collapse
|
30
|
Abstract
The perivascular adipose tissue (PVAT) has been recently recognized as an important factor in vascular biology, with implications in the pathogenesis of cardiovascular diseases. The cell types and the precursor cells of PVAT appear to be different according to their location, with the component cell type including white, brown, and beige adipocytes. PVAT releases a panel of adipokines and cytokines that maintain vascular homeostasis, but it also has the ability of intervention in the pathogenesis of the atherosclerotic plaques development and in the vascular tone modulation. In this review, we summarize the current knowledge and discuss the role of PVAT as a major contributing factor in the pathogenesis of ischemic coronary disease, hypertension, obesity, and diabetes mellitus. The new perspective of PVAT as an endocrine organ, along with the recent knowledge of the mechanisms involved in dysfunctional PVAT intervention in local vascular homeostasis perturbations, nowadays represent a new area of research in cardiovascular pathology, aiming to discover new therapeutic methods.
Collapse
|
31
|
A New Function for Perivascular Adipose Tissue (PVAT): Assistance of Arterial Stress Relaxation. Sci Rep 2020; 10:1807. [PMID: 32019956 PMCID: PMC7000722 DOI: 10.1038/s41598-020-58368-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 01/06/2020] [Indexed: 12/19/2022] Open
Abstract
In health, PVAT secretes anti-contractile factors that relax the underlying artery. PVAT’s contributions to vascular function include more than production of vasoactive substances. We hypothesized that PVAT benefits the artery by assisting the function of stress (–induced) relaxation. Thoracic aorta rings from Sprague Dawley rats were mounted in isolated tissue baths with (+) and without (−) PVAT. A cumulative length tension (0–6 grams) was generated. The tension to which the tissue stress relaxed over 30 minutes was recorded; the tension lost was stress relaxation. The presence of PVAT increased the amount of stress relaxation (final tension in mgs; aortic ring −PVAT = 4578 ± 190; aortic ring + PVAT = 2730 ± 274, p < 0.05). PVAT left attached but not encompassing the aorta provided no benefit in cumulative stress relaxation (aortic ring +/− PVAT = 4122 ± 176; p > 0.05 vs −PVAT). A PVAT ring separated from the aorta demonstrated more profound stress relaxation than did the aortic ring itself. Finally, PVAT-assisted stress relaxation was observed in an artery with white fat (superior mesenteric artery) and in aorta from both male and female of another rat strain, the Dahl S rat. Knowledge of this new PVAT function supports PVAT as an essential player in vascular health.
Collapse
|
32
|
Badran M, Yassin BA, Lin DTS, Kobor MS, Ayas N, Laher I. Gestational intermittent hypoxia induces endothelial dysfunction, reduces perivascular adiponectin and causes epigenetic changes in adult male offspring. J Physiol 2019; 597:5349-5364. [PMID: 31441069 DOI: 10.1113/jp277936] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/26/2019] [Indexed: 12/11/2022] Open
Abstract
KEY POINTS Obstructive sleep apnoea (OSA) is characterized by intermittent hypoxia, which causes oxidative stress and inflammation and increases the risk of cardiovascular disease. OSA during pregnancy causes adverse maternal and fetal outcomes. The effects of pre-existing OSA in pregnant women on cardiometabolic outcomes in the offspring are unknown. We evaluated basic metabolic parameters, as well as aortic vascular and perivascular adipose tissue (PVAT) function in response to adiponectin, and examined DNA methylation of adiponectin gene promoter in PVAT in 16-week-old adult offspring exposed to gestational intermittent hypoxia (GIH). GIH decreased body weights at week 1 in both male and female offspring, and caused subsequent increases in body weight and food consumption in male offspring only. Adult female offspring had normal levels of lipids, glucose and insulin, with no endothelial dysfunction. Adult male offspring exhibited dyslipidaemia, insulin resistance and hyperleptinaemia. Decreased endothelial-dependent vasodilatation, loss of anti-contractile activity of PVAT and low circulating PVAT adiponectin levels, as well as increased pro-inflammatory gene expression and DNA methylation of adiponectin gene promoter, occurred in adult male offspring. Our results suggest that male offspring of women with OSA could be at risk of developing cardiometabolic disease during adulthood. ABSTRACT Perturbations during pregnancy can program the offspring to develop cardiometabolic diseases later in life. Obstructive sleep apnoea (OSA) is a chronic condition that frequently affects pregnancies and leads to adverse fetal outcomes. We assessed the offspring of female mice experiencing gestational intermittent hypoxia (GIH), a hallmark of OSA, for changes in metabolic profiles, aortic nitric oxide (NO)-dependent relaxations, perivascular adipose tissue (PVAT) anti-contractile activities and the responses to adiponectin, and DNA methylation of the adiponectin gene promoter in PVAT tissue. Pregnant mouse dams were exposed to intermittent hypoxic cycles ( F I O 2 21-12%) for 18 days. GIH resulted in lower body weights of pups at week 1, followed by significant weight gain by week 16 of age in male but not female offspring. Plasma lipids, leptin and insulin resistance were higher in GIH male adult offspring. Endothelium-dependent relaxation in response to ACh and the anti-contractile activity of PVAT in the abdominal aorta was reduced in GIH adult male offspring. Incubation of arteries from GIH adult male offspring with adiponectin restored the anti-contractile activity of PVAT. Both circulating and PVAT tissue homogenate levels of adiponectin, as well as gene expression of adiponectin in PVAT, were lower in GIH male offspring, along with an increased gene expression of inflammatory cytokines. Pyrosequencing of adiponectin gene promoter in PVAT showed increased DNA methylation in GIH male offspring. Our results indicate that GIH leads to vascular disease in adult male offspring through PVAT dysfunction, which was associated with low adiponectin levels and epigenetic modifications on the adiponectin gene promoter.
Collapse
Affiliation(s)
- Mohammad Badran
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - Bisher Abu Yassin
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| | - David Tse Shen Lin
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Michael S Kobor
- Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, and Department of Medical Genetics, University of British Columbia, Vancouver, BC, Canada
| | - Najib Ayas
- Divisions of Critical Care and Respiratory Medicine, Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Sleep Disorders Program, UBC Hospital, Vancouver, BC, Canada.,Division of Critical Care Medicine, Providence Healthcare, Vancouver, BC, Canada
| | - Ismail Laher
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
33
|
Osikoya O, Ahmed H, Panahi S, Bourque SL, Goulopoulou S. Uterine perivascular adipose tissue is a novel mediator of uterine artery blood flow and reactivity in rat pregnancy. J Physiol 2019; 597:3833-3852. [PMID: 31165480 DOI: 10.1113/jp277643] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 05/30/2019] [Indexed: 12/12/2022] Open
Abstract
KEY POINTS In vivo, uterine perivascular adipose tissue (PVAT) potentiates uterine artery blood flow in pregnant rats, although not in non-pregnant rats. In isolated preparations, uterine PVAT has pro-contractile and anti-dilatory effects on uterine arteries. Pregnancy induces changes in uterine arteries that makes them responsive to uterine PVAT signalling. ABSTRACT An increase in uterine artery blood flow (UtBF) is a common and necessary feature of a healthy pregnancy. In the present study, we tested the hypothesis that adipose tissue surrounding uterine arteries (uterine perivascular adipose tissue; PVAT) is a novel local mediator of UtBF and uterine artery tone during pregnancy. In vivo experiments in anaesthetized Sprague-Dawley rats showed that pregnant animals (gestational day 16, term = 22--23 days) had a three-fold higher UtBF compared to non-pregnant animals. Surgical removal of uterine PVAT reduced UtBF only in pregnant rats. In a series of ex vivo bioassays, we demonstrated that uterine PVAT had pro-contractile and anti-dilatory effects on rat uterine arteries. In the presence of PVAT-conditioned media, isolated uterine arteries from both pregnant and non-pregnant rats had reduced vasodilatory responses. In non-pregnant rats, these responses were mediated at the level of uterine vascular smooth muscle, whereas, in pregnant rats, PVAT-media reduced endothelium-dependent relaxation. Pregnancy increased adipocyte size in ovarian adipose tissue but had no effect on uterine PVAT adipocyte morphology. In addition, pregnancy down-regulated the gene expression of metabolic adipokines in uterine but not in aortic PVAT. In conclusion, this is the first study to demonstrate that uterine PVAT plays a regulatory role in UtBF, at least in part, as a result of its actions on uterine artery tone. We propose that the interaction between the uterine vascular wall and its adjacent adipose tissue may provide new insights for interventions in pregnancies with adipose tissue dysfunction and abnormal UtBF.
Collapse
Affiliation(s)
- Oluwatobiloba Osikoya
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Hijab Ahmed
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Sareh Panahi
- Departments of Anesthesiology & Pain Medicine, Pharmacology, and Pediatrics, Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Stephane L Bourque
- Departments of Anesthesiology & Pain Medicine, Pharmacology, and Pediatrics, Women and Children's Health Research Institute, University of Alberta, Edmonton, AB, Canada
| | - Styliani Goulopoulou
- Department of Physiology and Anatomy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
34
|
Peterson SJ, Rubinstein R, Faroqui M, Raza A, Boumaza I, Zhang Y, Stec D, Abraham NG. Positive Effects of Heme Oxygenase Upregulation on Adiposity and Vascular Dysfunction: Gene Targeting vs. Pharmacologic Therapy. Int J Mol Sci 2019; 20:ijms20102514. [PMID: 31121826 PMCID: PMC6566770 DOI: 10.3390/ijms20102514] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Revised: 05/17/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022] Open
Abstract
Objective: Heme oxygenase (HO-1) plays a critical role in adipogenesis and it is important to understand its function in obesity. Many studies have shown that upregulation of HO-1 can affect the biologic parameters in obesity-mediated diabetes, hypertension and vascular endothelial cell function. Thus, we aimed to explore the hypothesis that upregulation of HO-1, using a pharmacologic approach as well as gene targeting, would improve both adiposity and endothelial cell dysfunction by direct targeting of endothelial cells. Our second aim was to compare the short-term effect of a HO-1 inducer, cobalt-protoporphrin IX (CoPP), with the long-term effects of gene targeted therapy on vascular and adipocyte stem cells in obese mice. Method: We examined the effect of CoPP on fat pre-adipocytes and mesenchymal stem cells (MSC) in mice fed a high-fat diet (HFD). We also used a lentiviral construct that expressed heme oxygenase (HO-1) that was under the control of an endothelium specific promoter, vascular endothelium cadherin (VECAD) heme oxygenase (VECAD-HO-1). We targeted endothelial cells using vascular endothelium cadherin/green fluorescent protein fusion construct (VECAD-GFP) as the control. Conditioned media (CM) from endothelial cells (EC) was added to fat derived adipocytes. Additionally, we treated renal interlobar arteries with phenylephrine and dosed cumulative increments of acetylcholine both with and without exposure to CoPP. We did the same vascular reactivity experiments with VECAD-HO-1 lentiviral construct compared to the control. Results: CoPP improved vascular reactivity and decreased adipogenesis compared to the control. MSCs exposed to CM from EC transfected with VECAD-HO-1 showed decreased adipogenesis, smaller lipid droplet size and decreased PPAR-γ, C/EBP and increased Wnt 10b compared to the control. HO-1 upregulation had a direct effect on reducing adipogenesis. This effect was blocked by tin mesoporphrin (SnMP). EC treated with VECAD-HO-1 expressed lower levels of ICAM and VCAM compared to the control, suggesting improved EC function. This also improved ACH induced vascular reactivity. These effects were also reversed by SnMP. The effect of viral transfection was much more specific and sustained than the effects of pharmacologic therapy, CoPP. Conclusion: This study demonstrates that a pharmacological inducer of HO-1 such as CoPP improves endothelial cell function while dampening adipogenesis, but long-term HO-1 expression by direct targeting of endothelial cells by gene transfer therapy may offer a more specific and ideal solution. This was evidenced by smaller healthier adipocytes that had improved insulin sensitivity, suggesting increased adiponectin levels. HO-1 upregulation reestablished the “crosstalk” between perivascular adipose tissue and the vascular system that was lost in the chronic inflammatory state of obesity. This study demonstrates that gene targeting of EC may well be the future direction in treating obesity induced EC dysfunction, with the finding that targeting the vasculature had a direct and sustained effect on adipogenesis.
Collapse
Affiliation(s)
- Stephen J Peterson
- Department of Medicine, Weill Cornell Medicine, New York, NY 10065, USA.
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA.
| | - Rochelle Rubinstein
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, USA.
| | - Mouzam Faroqui
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA.
| | - Adnan Raza
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA.
| | - Imene Boumaza
- New York Presbyterian Brooklyn Methodist Hospital, Brooklyn, NY 11215, USA.
| | - Yilun Zhang
- Tufts University School of Medicine, Boston, MA 02111, USA.
| | - David Stec
- Department of Physiology and Biophysics at the University of Mississippi Medical Center, Jackson, MI 39216, USA.
| | - Nader G Abraham
- Departments of Medicine and Pharmacology, New York Medical College, Valhalla, NY 10595, USA.
- Department of Pharmacology, Physiology and Toxicology, Marshall University, Joan Edwards School of Medicine, Huntington, WV 25701, USA.
| |
Collapse
|
35
|
Perivascular Adipose Tissue-Enhanced Vasodilation in Metabolic Syndrome Rats by Apelin and N-Acetyl⁻l-Cysteine-Sensitive Factor(s). Int J Mol Sci 2018; 20:ijms20010106. [PMID: 30597883 PMCID: PMC6337496 DOI: 10.3390/ijms20010106] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 12/24/2018] [Accepted: 12/24/2018] [Indexed: 01/24/2023] Open
Abstract
Perivascular adipose tissue (PVAT) can regulate vascular tone. In mesenteric arteries of SHRSP.Z-Leprfa/IzmDmcr rats (SHRSP.ZF) with metabolic syndrome, vascular dysfunction is compensated by PVAT-dependent mechanisms that disappear with increasing age. In this study, we investigated the mechanisms of the age-related changes and responsible factor(s) involved in the enhancing effects of mesenteric arterial PVAT in SHRSP.ZF. Acetylcholine- and sodium nitroprusside-induced relaxations of isolated arteries were greater with PVAT than without PVAT at 17 and 20 weeks of age (wks), and as expected, this enhancement by the presence of PVAT disappeared at 23 wks. PVAT mRNA levels of angiotensin II type 1 (AT1) receptor-associated protein was less and AT1 receptor was unchanged at 23 wks when compared to 20 wks. At 20 wks, the enhanced acetylcholine-induced relaxation by the presence of PVAT was inhibited by N-acetyl-l-cysteine (NAC). Acetylcholine-induced relaxation of arteries without PVAT was increased in the presence of exogenously added apelin. PVAT mRNA level of apelin was higher in SHRSP.ZF than in control Wistar-Kyoto rats, and the level was decreased with aging. These results suggest that AT1 receptor activation in PVAT, and changes in the regulation of apelin and a NAC-sensitive factor are related to the age-dependent deterioration of the vasodilation enhancing effects of mesenteric arterial PVAT in SHRSP.ZF.
Collapse
|
36
|
Byron KL, Brueggemann LI. Kv7 potassium channels as signal transduction intermediates in the control of microvascular tone. Microcirculation 2018; 25. [PMID: 28976052 DOI: 10.1111/micc.12419] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2017] [Accepted: 09/27/2017] [Indexed: 12/18/2022]
Abstract
Potassium channels are recognized as important regulators of cellular functions in most, if not all cell types. These cellular proteins assemble to form gated pores in the plasma membrane, which serve to regulate the flow of potassium ions (K+ ) from the cytosol to the extracellular space. In VSMCs, the open state of potassium channels enables the efflux of K+ and thereby establishes a negative resting voltage across the plasma membrane that inhibits the opening of VSCCs. Under these conditions, cytosolic Ca2+ concentrations are relatively low and Ca2+ -dependent contraction is inhibited. Recent research has identified Kv7 family potassium channels as important contributors to resting membrane voltage in VSMCs, with much of the research focusing on the effects of drugs that specifically activate or block these channels to produce corresponding effects on VSMC contraction and vascular tone. Increasingly, evidence is emerging that these channels are not just good drug targets-they are also essential intermediates in vascular signal transduction, mediating vasoconstrictor or vasodilator responses to a variety of physiological stimuli. This review will summarize recent research findings that support a crucial function of Kv7 channels in both positive (vasoconstrictive) and negative (vasorelaxant) regulation of microvascular tone.
Collapse
Affiliation(s)
- Kenneth L Byron
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| | - Lyubov I Brueggemann
- Department of Molecular Pharmacology & Therapeutics, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
37
|
Assersen KB, Jensen PS, Briones AM, Rasmussen LM, Marcussen N, Toft A, Vanhoutte PM, Jensen BL, Hansen PBL. Periarterial fat from two human vascular beds is not a source of aldosterone to promote vasoconstriction. Am J Physiol Renal Physiol 2018; 315:F1670-F1682. [PMID: 30280597 DOI: 10.1152/ajprenal.00391.2018] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Mouse adipocytes have been reported to release aldosterone and reduce endothelium-dependent relaxation. It is unknown whether perivascular adipose tissue (PVAT) releases aldosterone in humans. The present experiments were designed to test the hypothesis that human PVAT releases aldosterone and induces endothelial dysfunction. Vascular reactivity was assessed in human internal mammary and renal segmental arteries obtained at surgery. The arteries were prepared with/without PVAT, and changes in isometric tension were measured in response to the vasoconstrictor thromboxane prostanoid receptor agonist U46619 and the endothelium-dependent vasodilator acetylcholine. The effects of exogenous aldosterone and of mineralocorticoid receptor (MR) antagonist eplerenone were determined. Aldosterone concentrations were measured by ELISA in conditioned media incubated with human adipose tissue with/without angiotensin II stimulation. Presence of aldosterone synthase and MR mRNA was examined in perirenal, abdominal, and mammary PVAT by PCR. U46619 -induced tension and acetylcholine-induced relaxation were unaffected by exogenous and endogenous aldosterone (addition of aldosterone and MR blocker) in mammary and renal segmental arteries, both in the presence and absence of PVAT. Aldosterone release from incubated perivascular fat was not detectable. Aldosterone synthase expression was not consistently observed in human adipose tissues in contrast to that of MR. Thus, exogenous aldosterone does not affect vascular reactivity and endothelial function in ex vivo human arterial segments, and the tested human adipose tissues have no capacity to synthesize/release aldosterone. In perspective, physiologically relevant effects of aldosterone on vascular function in humans are caused by systemic aldosterone originating from the adrenal gland.
Collapse
Affiliation(s)
- Kasper B Assersen
- Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark
| | - Pia S Jensen
- Department for Clinical Biochemistry and Pharmacology, Center for Individualized Medicine in Arterial Disease, Odense University Hospital , Odense , Denmark
| | - Ana M Briones
- Departamento de Farmacología, Universidad Autónoma de Madrid, Instituto de Investigación Hospital La Paz, Centro de Investigación en Red en Enfermedades Cardiovasculares , Madrid , Spain
| | - Lars M Rasmussen
- Department for Clinical Biochemistry and Pharmacology, Center for Individualized Medicine in Arterial Disease, Odense University Hospital , Odense , Denmark
| | - Niels Marcussen
- Department of Pathology, Odense University Hospital , Odense , Denmark
| | - Anja Toft
- Department of Urology, Odense University Hospital , Odense , Denmark
| | - Paul M Vanhoutte
- State Key Laboratory of Pharmaceutical Biotechnology and Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong, China
| | - Boye L Jensen
- Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark
| | - Pernille B L Hansen
- Cardiovascular and Renal Research, University of Southern Denmark , Odense , Denmark.,Cardiovascular, Renal and Metabolic, Innovative Medicines and Early Development Biotech Unit, AstraZeneca, Gothenburg , Sweden
| |
Collapse
|
38
|
Martinez-Quinones P, McCarthy CG, Watts SW, Klee NS, Komic A, Calmasini FB, Priviero F, Warner A, Chenghao Y, Wenceslau CF. Hypertension Induced Morphological and Physiological Changes in Cells of the Arterial Wall. Am J Hypertens 2018; 31:1067-1078. [PMID: 29788246 DOI: 10.1093/ajh/hpy083] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/14/2018] [Indexed: 12/17/2022] Open
Abstract
Morphological and physiological changes in the vasculature have been described in the evolution and maintenance of hypertension. Hypertension-induced vascular dysfunction may present itself as a contributing, or consequential factor, to vascular remodeling caused by chronically elevated systemic arterial blood pressure. Changes in all vessel layers, from the endothelium to the perivascular adipose tissue (PVAT), have been described. This mini-review focuses on the current knowledge of the structure and function of the vessel layers, specifically muscular arteries: intima, media, adventitia, PVAT, and the cell types harbored within each vessel layer. The contributions of each cell type to vessel homeostasis and pathophysiological development of hypertension will be highlighted.
Collapse
Affiliation(s)
- Patricia Martinez-Quinones
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Cameron G McCarthy
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Nicole S Klee
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Amel Komic
- Department of Surgery, Medical College of Georgia at Augusta University, Augusta, Georgia
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Fabiano B Calmasini
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Fernanda Priviero
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Alexander Warner
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Yu Chenghao
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| | - Camilla F Wenceslau
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, Georgia
| |
Collapse
|
39
|
Rivera-Mancía S, Colín-Ramírez E, Cartas-Rosado R, Infante O, Vargas-Barrón J, Vallejo M. Indicators of accumulated fat are stronger associated with prehypertension compared with indicators of circulating fat: A cross-sectional study. Medicine (Baltimore) 2018; 97:e11869. [PMID: 30142781 PMCID: PMC6113050 DOI: 10.1097/md.0000000000011869] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Recently, prehypertension has been considered as a risk factor for cardiovascular disease because it can progress to hypertension. The association between obesity and dyslipidemia with raised blood pressure has been reported in some studies; however, the ability of indicators of such conditions to predict prehypertension has been scarcely explored. In this cross-sectional study, we compared the ability of indicators of accumulated and circulating fat to discriminate between prehypertensive and normotensive Mexico City residents (n = 1377). The indicators were classified based on the parameters needed for their calculation: including only circulating fat (IOCFi) (e.g., Castelli risk indexes), including only accumulated fat (IOAFi) (e.g., waist circumference [WC]), and mixed (e.g., lipid accumulation product [LAP]). We compared the areas under the receiving operating characteristic curves (AURCs) and estimated the cutoff points for each indicator and their associated risk of prehypertension. The IOAFi had the greatest AURCs, followed by mixed and IOCFi; the AURCs for WC were the highest (AURC = 0.688 and 0.666 for women and men, respectively). The highest odds ratios for prehypertension were those associated with the cutoff points for IOAFi and LAP (e.g., OR = 2.8 for women with WC > 83.5 cm and OR = 2.6 for men with WC > 87.5 cm). Early detecting people at risk of cardiovascular disease is a necessity and given that WC had a better performance than the other indexes and it is relatively easy to measure, it has the potential of being used as a complementary measure in routine clinical examinations and by the general population as an auto-screening measurement to detect prehypertension.
Collapse
Affiliation(s)
| | | | | | | | | | - Maite Vallejo
- Department of Social Medicine Research, National Institute of Cardiology “Ignacio Chávez,” Mexico City, Mexico
| |
Collapse
|
40
|
Chang L, Xiong W, Zhao X, Fan Y, Guo Y, Garcia-Barrio M, Zhang J, Jiang Z, Lin JD, Chen YE. Bmal1 in Perivascular Adipose Tissue Regulates Resting-Phase Blood Pressure Through Transcriptional Regulation of Angiotensinogen. Circulation 2018; 138:67-79. [PMID: 29371216 PMCID: PMC6030431 DOI: 10.1161/circulationaha.117.029972] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Accepted: 01/16/2018] [Indexed: 02/07/2023]
Abstract
BACKGROUND The perivascular adipose tissue (PVAT) surrounding vessels constitutes a distinct functional integral layer of the vasculature required to preserve vascular tone under physiological conditions. However, there is little information on the relationship between PVAT and blood pressure regulation, including its potential contributions to circadian blood pressure variation. METHODS Using unique brown adipocyte-specific aryl hydrocarbon receptor nuclear translocator-like protein 1 (Bmal1) and angiotensinogen knockout mice, we determined the vasoactivity of homogenized PVAT in aortic rings and how brown adipocyte peripheral expression of Bmal1 and angiotensinogen in PVAT regulates the amplitude of diurnal change in blood pressure in mice. RESULTS We uncovered a peripheral clock in PVAT and demonstrated that loss of Bmal1 in PVAT reduces blood pressure in mice during the resting phase, leading to a superdipper phenotype. PVAT extracts from wild-type mice significantly induced contractility of isolated aortic rings in vitro in an endothelium-independent manner. This property was impaired in PVAT from brown adipocyte-selective Bmal1-deficient (BA-Bmal1-KO) mice. The PVAT contractile properties were mediated by local angiotensin II, operating through angiotensin II type 1 receptor-dependent signaling in the isolated vessels and linked to PVAT circadian regulation of angiotensinogen. Indeed, angiotensinogen mRNA and angiotensin II levels in PVAT of BA-Bmal1-KO mice were significantly reduced. Systemic infusion of angiotensin II, in turn, reduced Bmal1 expression in PVAT while eliminating the hypotensive phenotype during the resting phase in BA-Bmal1-KO mice. Angiotensinogen, highly expressed in PVAT, shows circadian expression in PVAT, and selective deletion of angiotensinogen in brown adipocytes recapitulates the phenotype of selective deletion of Bmal1 in brown adipocytes. Furthermore, angiotensinogen is a transcriptional target of Bmal1 in PVAT. CONCLUSIONS These data indicate that local Bmal1 in PVAT regulates angiotensinogen expression and the ensuing increase in angiotensin II, which acts on smooth muscle cells in the vessel walls to regulate vasoactivity and blood pressure in a circadian fashion during the resting phase. These findings will contribute to a better understanding of the cardiovascular complications of circadian disorders, alterations in the circadian dipping phenotype, and cross-talk between systemic and peripheral regulation of blood pressure.
Collapse
Affiliation(s)
- Lin Chang
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| | - Wenhao Xiong
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang (W.X., Z.J.)
| | - Xiangjie Zhao
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| | - Yanbo Fan
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| | - Yanhong Guo
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| | - Minerva Garcia-Barrio
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| | - Jifeng Zhang
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| | - Zhisheng Jiang
- Institute of Cardiovascular Disease, Key Laboratory for Atherosclerology of Hunan Province, University of South China, Hengyang (W.X., Z.J.)
| | - Jiandie D Lin
- Life Sciences Institute (J.D.L.)
- Department of Cell and Developmental Biology (J.D.L.), University of Michigan, Ann Arbor
| | - Y Eugene Chen
- Cardiovascular Center, Department of Internal Medicine (L.C., X.Z., Y.F., Y.G., M.G.B., J.Z., Y.E.C.)
| |
Collapse
|
41
|
Kennedy S, Salt IP. Molecular mechanisms regulating perivascular adipose tissue - potential pharmacological targets? Br J Pharmacol 2018; 174:3385-3387. [PMID: 28940457 DOI: 10.1111/bph.13969] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, Glasgow, UK
| |
Collapse
|
42
|
Prado NJ, Ferder L, Manucha W, Diez ER. Anti-Inflammatory Effects of Melatonin in Obesity and Hypertension. Curr Hypertens Rep 2018; 20:45. [PMID: 29744660 DOI: 10.1007/s11906-018-0842-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Here, we review the known relations between hypertension and obesity to inflammation and postulate the endogenous protective effect of melatonin and its potential as a therapeutic agent. We will describe the multiple effects of melatonin on blood pressure, adiposity, body weight, and focus on mitochondrial-related anti-inflammatory and antioxidant protective effects. RECENT FINDINGS Hypertension and obesity are usually associated with systemic and tissular inflammation. The progressive affection of target-organs involves multiple mediators of inflammation, most of them redundant, which make anti-inflammatory strategies ineffective. Melatonin reduces blood pressure, body weight, and inflammation. The mechanisms of action of this ancient molecule of protection involve multiple levels of action, from subcellular to intercellular. Mitochondria is a key inflammatory element in vascular and adipose tissue and a potential pharmacological target. Melatonin protects against mitochondrial dysfunction. Melatonin reduces blood pressure and adipose tissue dysfunction by multiple anti-inflammatory/antioxidant actions and provides potent protection against mitochondria-mediated injury in hypertension and obesity. This inexpensive and multitarget molecule has great therapeutic potential against both epidemic diseases.
Collapse
Affiliation(s)
- Natalia Jorgelina Prado
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina
| | - León Ferder
- Pediatric Department Nephrology Division, Miller School of Medicine, University of Miami, Florida, USA
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina.,Área de Farmacología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Emiliano Raúl Diez
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Mendoza, Argentina. .,Instituto de Fisiología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, Centro Universitario, CP 5500, Mendoza, Argentina.
| |
Collapse
|
43
|
Bäckdahl J, Rydén M. Bariatric surgery helps to reduce blood pressure - insights from GATEWAY trial. Cardiovasc Res 2018; 114:e19-e21. [PMID: 29481648 DOI: 10.1093/cvr/cvy008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Jesper Bäckdahl
- Department of Medicine (H7), C2-94, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| | - Mikael Rydén
- Department of Medicine (H7), C2-94, Karolinska Institutet, Karolinska University Hospital, Huddinge, 141 86 Stockholm, Sweden
| |
Collapse
|
44
|
Roostalu U, Wong JK. Arterial smooth muscle dynamics in development and repair. Dev Biol 2018; 435:109-121. [PMID: 29397877 DOI: 10.1016/j.ydbio.2018.01.018] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Revised: 01/08/2018] [Accepted: 01/24/2018] [Indexed: 12/11/2022]
Abstract
Arterial vasculature distributes blood from early embryonic development and provides a nutrient highway to maintain tissue viability. Atherosclerosis, peripheral artery diseases, stroke and aortic aneurysm represent the most frequent causes of death and are all directly related to abnormalities in the function of arteries. Vascular intervention techniques have been established for the treatment of all of these pathologies, yet arterial surgery can itself lead to biological changes in which uncontrolled arterial wall cell proliferation leads to restricted blood flow. In this review we describe the intricate cellular composition of arteries, demonstrating how a variety of distinct cell types in the vascular walls regulate the function of arteries. We provide an overview of the developmental origin of arteries and perivascular cells and focus on cellular dynamics in arterial repair. We summarize the current knowledge of the molecular signaling pathways that regulate vascular smooth muscle differentiation in the embryo and in arterial injury response. Our review aims to highlight the similarities as well as differences between cellular and molecular mechanisms that control arterial development and repair.
Collapse
Affiliation(s)
- Urmas Roostalu
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK.
| | - Jason Kf Wong
- Manchester Academic Health Science Centre, Division of Cell Matrix Biology and Regenerative Medicine, Faculty of Biology, Medicine and Health, School of Biological Sciences, University of Manchester, UK; Department of Plastic Surgery, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK.
| |
Collapse
|
45
|
Tumor necrosis factor-α receptor 1 contributes to ethanol-induced vascular reactive oxygen species generation and hypertension. ACTA ACUST UNITED AC 2017; 11:684-696.e3. [DOI: 10.1016/j.jash.2017.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/22/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023]
|
46
|
Ramirez JG, O'Malley EJ, Ho WSV. Pro-contractile effects of perivascular fat in health and disease. Br J Pharmacol 2017; 174:3482-3495. [PMID: 28257140 DOI: 10.1111/bph.13767] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Revised: 02/23/2017] [Accepted: 02/23/2017] [Indexed: 12/28/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is now recognized as an active player in vascular homeostasis. The expansion of PVAT in obesity and its possible role in vascular dysfunction have attracted much interest. In terms of the regulation of vascular tone and blood pressure, PVAT has been shown to release vasoactive mediators, for instance, angiotensin peptides, reactive oxygen species, chemokines and cytokines. The secretory profile of PVAT is altered by obesity, hypertension and other cardiovascular diseases, leading to an imbalance between its pro-contractile and anti-contractile effects. PVAT adipocytes represent an important source of the mediators, but infiltrating immune cells may become more important under conditions of hypoxia and inflammation. This review describes recent advances in the effects of PVAT on the regulation of vascular tone, highlighting the evidence for a pro-contractile action in health and disease. The role of the endothelium, vascular smooth muscle, immune cells and probably perivascular nerves in PVAT function is also discussed. LINKED ARTICLES This article is part of a themed section on Molecular Mechanisms Regulating Perivascular Adipose Tissue - Potential Pharmacological Targets? To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.20/issuetoc.
Collapse
Affiliation(s)
- J G Ramirez
- Vascular Biology Research Centre, St George's University of London, London, SW17 0RE, UK
| | - E J O'Malley
- Vascular Biology Research Centre, St George's University of London, London, SW17 0RE, UK
| | - W S V Ho
- Vascular Biology Research Centre, St George's University of London, London, SW17 0RE, UK
| |
Collapse
|