1
|
Hwej A, Al-Ferjani A, Alshuweishi Y, Naji A, Kennedy S, Salt IP. Lack of AMP-activated protein kinase-α1 reduces nitric oxide synthesis in thoracic aorta perivascular adipose tissue. Vascul Pharmacol 2024; 157:107437. [PMID: 39433170 DOI: 10.1016/j.vph.2024.107437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/23/2024] [Accepted: 10/18/2024] [Indexed: 10/23/2024]
Abstract
OBJECTIVE Perivascular adipose tissue (PVAT) releases anti-contractile bioactive molecules including NO. PVAT anti-contractile activity is attenuated in mice lacking AMPKα1 (AMP-activated protein kinase-α1). As AMPK regulates endothelial NO synthase (eNOS) activity in cultured cells, NO synthesis was examined in PVAT from AMPKα1 knockout (KO) mice. METHODS AND RESULTS Endothelium-denuded thoracic or abdominal aortic rings were isolated from wild type (WT) and KO mice. NOS inhibition enhanced vasoconstriction in PVAT-intact thoracic aortic rings from mice of either genotype yet had no effect on abdominal rings as assessed by wire myography. Thoracic aorta PVAT exhibited increased NO production, NOS activity and levels of the brown adipose tissue marker uncoupling protein-1 (UCP1) compared to abdominal PVAT. In KO mice, NO production was significantly reduced in thoracic but not abdominal PVAT. Reduced NO production in KO thoracic PVAT was not due to altered levels or phosphorylation of eNOS but was associated with increased caveolin-1:eNOS association and caveolin-1 Tyr14 phosphorylation. A peptide that disrupts eNOS:caveolin-1 association increased NO synthesis and reduced vasoconstriction of PVAT-intact thoracic but not abdominal aortic rings. KO thoracic PVAT also exhibited reduced UCP1 levels. CONCLUSIONS Murine thoracic aorta PVAT exhibits higher NO synthesis and UCP1 levels than abdominal aortic PVAT. Downregulation of AMPK suppresses NO synthesis which may contribute to the reduced anticontractile activity and reduced brown adipose tissue phenotype of KO thoracic PVAT. The mechanism underlying the effect of AMPK downregulation likely results from increased caveolin-1:eNOS association associated with caveolin-1 Tyr14 phosphorylation.
Collapse
Affiliation(s)
- Abdmajid Hwej
- School of Cardiovascular and Metabolic Health, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, United Kingdom; School of Pharmacy, University of El-Mergib, Al-Khoms, Libya
| | - Ali Al-Ferjani
- School of Cardiovascular and Metabolic Health, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Faculty of Medicine, University of Tripoli, Tripoli, Libya
| | - Yazeed Alshuweishi
- School of Molecular Biosciences, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Clinical Laboratory Sciences, King Saud University, Riyadh 12372, Saudi Arabia
| | - Abdullah Naji
- School of Cardiovascular and Metabolic Health, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, United Kingdom; School of Pharmacy, Department of Pharmacology, Najran University, Najran, Saudi Arabia
| | - Simon Kennedy
- School of Cardiovascular and Metabolic Health, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ian P Salt
- School of Molecular Biosciences, College of Veterinary, Medical and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| |
Collapse
|
2
|
Sigdel S, Udoh G, Albalawy R, Wang J. Perivascular Adipose Tissue and Perivascular Adipose Tissue-Derived Extracellular Vesicles: New Insights in Vascular Disease. Cells 2024; 13:1309. [PMID: 39195199 PMCID: PMC11353161 DOI: 10.3390/cells13161309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 07/29/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
Perivascular adipose tissue (PVAT) is a special deposit of fat tissue surrounding the vasculature. Previous studies suggest that PVAT modulates the vasculature function in physiological conditions and is implicated in the pathogenesis of vascular diseases. Understanding how PVAT influences vasculature function and vascular disease progression is important. Extracellular vesicles (EVs) are novel mediators of intercellular communication. EVs encapsulate molecular cargo such as proteins, lipids, and nucleic acids. EVs can influence cellular functions by transferring the carried bioactive molecules. Emerging evidence indicates that PVAT-derived EVs play an important role in vascular functions under health and disease conditions. This review will focus on the roles of PVAT and PVAT-EVs in obesity, diabetic, and metabolic syndrome-related vascular diseases, offering novel insights into therapeutic targets for vascular diseases.
Collapse
Affiliation(s)
- Smara Sigdel
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (S.S.); (G.U.)
| | - Gideon Udoh
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (S.S.); (G.U.)
| | - Rakan Albalawy
- Department of Internal Medicine, Joan C Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA;
| | - Jinju Wang
- Department of Biomedical Sciences, Joan C Edwards School of Medicine, Marshall University, Huntington, WV 25755, USA; (S.S.); (G.U.)
| |
Collapse
|
3
|
Hanscom M, Morales-Soto W, Watts SW, Jackson WF, Gulbransen BD. Innervation of adipocytes is limited in mouse perivascular adipose tissue. Am J Physiol Heart Circ Physiol 2024; 327:H155-H181. [PMID: 38787382 PMCID: PMC11380956 DOI: 10.1152/ajpheart.00041.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/10/2024] [Accepted: 05/14/2024] [Indexed: 05/25/2024]
Abstract
Perivascular adipose tissue (PVAT) regulates vascular tone by releasing anticontractile factors. These anticontractile factors are driven by processes downstream of adipocyte stimulation by norepinephrine; however, whether norepinephrine originates from neural innervation or other sources is unknown. The goal of this study was to test the hypothesis that neurons innervating PVAT provide the adrenergic drive to stimulate adipocytes in aortic and mesenteric perivascular adipose tissue (aPVAT and mPVAT), and white adipose tissue (WAT). Healthy male and female mice (8-13 wk) were used in all experiments. Expression of genes associated with synaptic transmission were quantified by qPCR and adipocyte activity in response to neurotransmitters and neuron depolarization was assessed in AdipoqCre+;GCaMP5g-tdTf/WT mice. Immunostaining, tissue clearing, and transgenic reporter lines were used to assess anatomical relationships between nerves and adipocytes. Although synaptic transmission component genes are expressed in adipose tissues (aPVAT, mPVAT, and WAT), strong nerve stimulation with electrical field stimulation does not significantly trigger calcium responses in adipocytes. However, norepinephrine consistently elicits strong calcium responses in adipocytes from all adipose tissues studied. Bethanechol induces minimal adipocyte responses. Imaging neural innervation using various techniques reveals that nerve fibers primarily run alongside blood vessels and rarely branch into the adipose tissue. Although nerve fibers are associated with blood vessels in adipose tissue, they demonstrate limited anatomical and functional interactions with adjacent adipocytes, challenging the concept of classical innervation. These findings dispute the significant involvement of neural input in regulating PVAT adipocyte function and emphasize alternative mechanisms governing adrenergic-driven anticontractile functions of PVAT.NEW & NOTEWORTHY This study challenges prevailing views on neural innervation in perivascular adipose tissue (PVAT) and its role in adrenergic-driven anticontractile effects on vasculature. Contrary to existing paradigms, limited anatomical and functional connections were found between PVAT nerve fibers and adipocytes, underscoring the importance of exploring alternative mechanistic pathways. Understanding the mechanisms involved in PVAT's anticontractile effects is critical for developing potential therapeutic interventions against dysregulated vascular tone, hypertension, and cardiovascular disease.
Collapse
Affiliation(s)
- Marie Hanscom
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| | - Wilmarie Morales-Soto
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota, United States
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - William F Jackson
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, Michigan, United States
| | - Brian D Gulbransen
- Department of Physiology, Michigan State University, East Lansing, Michigan, United States
| |
Collapse
|
4
|
Tsvetkov D, Schleifenbaum J, Wang Y, Kassmann M, Polovitskaya MM, Ali M, Schütze S, Rothe M, Luft FC, Jentsch TJ, Gollasch M. KCNQ5 Controls Perivascular Adipose Tissue-Mediated Vasodilation. Hypertension 2024; 81:561-571. [PMID: 38354270 DOI: 10.1161/hypertensionaha.123.21834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 12/12/2023] [Indexed: 02/16/2024]
Abstract
BACKGROUND Small arteries exhibit resting tone, a partially contracted state that maintains arterial blood pressure. In arterial smooth muscle cells, potassium channels control contraction and relaxation. Perivascular adipose tissue (PVAT) has been shown to exert anticontractile effects on the blood vessels. However, the mechanisms by which PVAT signals small arteries, and their relevance remain largely unknown. We aimed to uncover key molecular components in adipose-vascular coupling. METHODS A wide spectrum of genetic mouse models targeting Kcnq3, Kcnq4, and Kcnq5 genes (Kcnq3-/-, Kcnq4-/-, Kcnq5-/-, Kcnq5dn/dn, Kcnq4-/-/Kcnq5dn/dn, and Kcnq4-/-/Kcnq5-/-), telemetry blood pressure measurements, targeted lipidomics, RNA-Seq profiling, wire-myography, patch-clamp, and sharp-electrode membrane potential measurements was used. RESULTS We show that PVAT causes smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels to hyperpolarize the membrane potential. This effect relaxes small arteries and regulates blood pressure. Oxygenation of polyunsaturated fats generates oxylipins, a superclass of lipid mediators. We identified numerous oxylipins released by PVAT, which potentiate vasodilatory action in small arteries by opening smooth muscle cell KV7.5 family of voltage-gated potassium (K+) channels. CONCLUSIONS Our results reveal a key molecular function of the KV7.5 family of voltage-gated potassium (K+) channels in the adipose-vascular coupling, translating PVAT signals, particularly oxylipins, to the central physiological function of vasoregulation. This novel pathway opens new therapeutic perspectives.
Collapse
Affiliation(s)
- Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| | - Johanna Schleifenbaum
- Institute of Vegetative Physiology, Charité-Universitätsmedizin Berlin, Germany (J.S.)
| | - Yibin Wang
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany (Y.W., F.C.L.)
| | - Mario Kassmann
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| | - Maya M Polovitskaya
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (M.M.P., S.S., T.J.J.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (M.M.P., S.S., T.J.J.)
| | - Mohamed Ali
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| | - Sebastian Schütze
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (M.M.P., S.S., T.J.J.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (M.M.P., S.S., T.J.J.)
| | | | - Friedrich C Luft
- Experimental and Clinical Research Center, a joint cooperation between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine, Berlin, Germany (Y.W., F.C.L.)
| | - Thomas J Jentsch
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie, Berlin, Germany (M.M.P., S.S., T.J.J.)
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany (M.M.P., S.S., T.J.J.)
- NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Berlin, Germany (T.J.J.)
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Germany (D.T., M.K., M.A., M.G.)
| |
Collapse
|
5
|
Jüttner AA, Ataei Ataabadi E, Golshiri K, de Vries R, Garrelds IM, Danser AHJ, Visser JA, Roks AJM. Adiponectin secretion by perivascular adipose tissue supports impaired vasodilation in a mouse model of accelerated vascular smooth muscle cell and adipose tissue aging. Vascul Pharmacol 2024; 154:107281. [PMID: 38320678 DOI: 10.1016/j.vph.2024.107281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 01/26/2024] [Accepted: 02/02/2024] [Indexed: 02/08/2024]
Abstract
OBJECTIVE Perivascular adipose tissue (PVAT) function during aging has not been investigated in detail so far and its effect on vasodilation remains to be fully elucidated. The aim of this study was to investigate endothelium-dependent vasodilation of thoracic aorta in a mouse model of accelerated, selective vascular smooth muscle and PVAT aging, induced by SM22α-Cre-driven genetic deletion of the endonuclease ERCC1 (SMC-KO mice) versus healthy littermates (LM). We hypothesized that PVAT enhances vasodilation in LM, possibly through adiponectin secretion, which might be compromised in SMC-KO animals. METHODS Thoracic aorta was isolated from SMC-KO animals and LM and segments with and without PVAT were mounted in wire myography setups. The endothelium-dependent vasodilation was assessed via acetylcholine dose-response curves and pathway contribution was studied. Moreover, adiponectin secretion was measured after stimulating the aortic segments with PVAT with acetylcholine. RESULTS Adiponectin, secreted by PVAT, led to increased NO-contribution to endothelium-dependent vasodilation in healthy LM, although this did not increase maximum relaxation due to loss of EDH. Endothelium-dependent vasodilation was decreased in SMC-KO animals due to reduced NO-contribution and complete EDH loss. Despite strong lipodystrophy the PVAT partially compensated for lost vasodilation in SMC-KO. LM PVAT contained acetylcholinesterase that attenuated acetylcholine responses. This was lost in SMC-KO. CONCLUSIONS PVAT-derived adiponectin is able to partially compensate for age-related decline in NO-mediated vasodilation, even during strong lipodystrophy, in conditions of absence of compensating EDH. In aorta with healthy PVAT acetylcholinesterase modulates vascular tone, but this is lost during aging, further compensating for decreased acetylcholine responsiveness. Thus, preservation of adiponectin levels, through relatively increased production in lipodystrophic PVAT, and reduction of cholinesterase might be regulatory mechanisms of the PVAT to preserve cholinergic vasodilation during aging.
Collapse
Affiliation(s)
- A A Jüttner
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - E Ataei Ataabadi
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - K Golshiri
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - R de Vries
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - I M Garrelds
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - A H J Danser
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - J A Visser
- Division of Endocrinology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| | - A J M Roks
- Division of Vascular Medicine and Pharmacology, Department of Internal Medicine, Erasmus MC, University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands.
| |
Collapse
|
6
|
Man AWC, Zhou Y, Reifenberg G, Camp A, Münzel T, Daiber A, Xia N, Li H. Deletion of adipocyte NOS3 potentiates high-fat diet-induced hypertension and vascular remodelling via chemerin. Cardiovasc Res 2023; 119:2755-2769. [PMID: 37897505 PMCID: PMC10757584 DOI: 10.1093/cvr/cvad164] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 07/17/2023] [Accepted: 10/27/2023] [Indexed: 10/30/2023] Open
Abstract
AIMS Obesity is an epidemic that is a critical contributor to hypertension and other cardiovascular diseases. Current paradigms suggest that endothelial nitric oxide synthase (eNOS/NOS3) in the vessel wall is the primary regulator of vascular function and blood pressure. However, recent studies have revealed the presence of eNOS/NOS3 in the adipocytes of white adipose tissues and perivascular adipose tissues (PVATs). The current understanding of the role of adipocyte NOS3 is based mainly on studies using global knockout models. The present study aimed to elucidate the functional significance of adipocyte NOS3 for vascular function and blood pressure control. METHODS AND RESULTS We generated an adipocyte-specific NOS3 knockout mouse line using adiponectin promoter-specific Cre-induced gene inactivation. Control and adipocyte-specific NOS3 knockout (A-NOS3 KO) mice were fed a high-fat diet (HFD). Despite less weight gain, A-NOS3 KO mice exhibited a significant increase in blood pressure after HFD feeding, associated with exacerbated vascular dysfunction and remodelling. A-NOS3 KO mice also showed increased expression of signature markers of inflammation and hypoxia in the PVATs. Among the differentially expressed adipokines, we have observed an upregulation of a novel adipokine, chemerin, in A-NOS3 KO mice. Chemerin was recently reported to link obesity and vascular dysfunction. Treatment with chemerin neutralizing antibody normalized the expression of remodelling markers in the aorta segments cultured in serum from HFD-fed A-NOS3 KO mice ex vivo. CONCLUSION These data suggest that NOS3 in adipocytes is vital in maintaining vascular homeostasis; dysfunction of adipocyte NOS3 contributes to obesity-induced vascular remodelling and hypertension.
Collapse
Affiliation(s)
- Andy W C Man
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Yawen Zhou
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Gisela Reifenberg
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Alica Camp
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Thomas Münzel
- Department of Cardiology, Cardiology I, Johannes Gutenberg University Medical Center, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Andreas Daiber
- Department of Cardiology, Cardiology I, Johannes Gutenberg University Medical Center, Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| | - Ning Xia
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
| | - Huige Li
- Department of Pharmacology, Johannes Gutenberg University Medical Center, Langenbeckstr. 1, 55131 Mainz, Germany
- German Centre for Cardiovascular Research (DZHK), Partner Site Rhine-Main, Mainz, Germany
| |
Collapse
|
7
|
Abstract
Obesity, which has currently reached pandemic dimensions, is usually accompanied by diabetes mellitus type 2 (T2DM). These two conditions share common pathophysiological mechanisms. Adipose tissue secretes cytokines which are involved in inflammation and various endocrine functions. As for T2DM, it is characterized also by inflammation, mitochondrial dysfunction, and hyperinsulinemia. These conditions occur also in other diseases related to obesity and T2DM, like cardiovascular disease (CVD) and nonalcoholic fatty liver disease (NAFLD). Thus, management of obesity-related complications with lifestyle modification, anti-obesity drugs, and bariatric surgery, all contribute to improvement in any of these conditions. This review provides an overview of the literature addressing the association between obesity and T2DM, briefly discussing the pathophysiological mechanisms linking these conditions and outlining the management approach at the overlap of obesity and T2DM.
Collapse
Affiliation(s)
- Chrysoula Boutari
- Department of Internal Medicine, Medical School, Aristotle University of Thessaloniki, Hippokration General Hospital, Thessaloniki, Greece; Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Antea DeMarsilis
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Christos S Mantzoros
- Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA; Department of Medicine, Boston VA Healthcare System, Boston, MA, USA.
| |
Collapse
|
8
|
Adachi Y, Ueda K, Takimoto E. Perivascular adipose tissue in vascular pathologies-a novel therapeutic target for atherosclerotic disease? Front Cardiovasc Med 2023; 10:1151717. [PMID: 37304960 PMCID: PMC10250715 DOI: 10.3389/fcvm.2023.1151717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/28/2023] [Indexed: 06/13/2023] Open
Abstract
Most blood vessels are surrounded by adipose tissues known as perivascular adipose tissue (PVAT). Emerging experimental data have implicated the potential involvement of PVAT in the pathogenesis of cardiovascular disease: PVAT might be a source of inflammatory mediators under pathological conditions such as metabolic disorders, chronic inflammation, and aging, leading to vascular pathologies, while having vasculo-protective roles in a healthy state. PVAT has been also gaining attention in human disease conditions. Recent integrative omics approaches have greatly enhanced our understanding of the molecular mechanisms underlying the diverse functions of PVAT. This review summarizes recent progress in PVAT research and discusses the potential of PVAT as a target for the treatment of atherosclerosis.
Collapse
|
9
|
Huang CL, Huang YN, Yao L, Li JP, Zhang ZH, Huang ZQ, Chen SX, Zhang YL, Wang JF, Chen YX, Liu ZY. Thoracic perivascular adipose tissue inhibits VSMC apoptosis and aortic aneurysm formation in mice via the secretome of browning adipocytes. Acta Pharmacol Sin 2023; 44:345-355. [PMID: 35945313 PMCID: PMC9889802 DOI: 10.1038/s41401-022-00959-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 07/12/2022] [Indexed: 02/04/2023]
Abstract
Abdominal aortic aneurysm (AAA) is a dangerous vascular disease without any effective drug therapies so far. Emerging evidence suggests the phenotypic differences in perivascular adipose tissue (PVAT) between regions of the aorta are implicated in the development of atherosclerosis evidenced by the abdominal aorta more vulnerable to atherosclerosis than the thoracic aorta in large animals and humans. The prevalence of thoracic aortic aneurysms (TAA) is much less than that of abdominal aortic aneurysms (AAA). In this study we investigated the effect of thoracic PVAT (T-PVAT) transplantation on aortic aneurysm formation and the impact of T-PVAT on vascular smooth muscle cells. Calcium phosphate-induced mouse AAA model was established. T-PVAT (20 mg) was implanted around the abdominal aorta of recipient mice after removal of endogenous abdominal PVAT (A-PVAT) and calcium phosphate treatment. Mice were sacrificed two weeks after the surgery and the maximum external diameter of infrarenal aorta was measured. We found that T-PVAT displayed a more BAT-like phenotype than A-PVAT; transplantation of T-PVAT significantly attenuated calcium phosphate-induced abdominal aortic dilation and elastic degradation as compared to sham control or A-PVAT transplantation. In addition, T-PVAT transplantation largely preserved smooth muscle cell content in the abdominal aortic wall. Co-culture of T-PVAT with vascular smooth muscle cells (VSMCs) significantly inhibited H2O2- or TNFα plus cycloheximide-induced VSMC apoptosis. RNA sequencing analysis showed that T-PVAT was enriched by browning adipocytes and anti-apoptotic secretory proteins. We further verified that the secretome of mature adipocytes isolated from T-PVAT significantly inhibited H2O2- or TNFα plus cycloheximide-induced VSMC apoptosis. Using proteomic and bioinformatic analyses we identified cartilage oligomeric matrix protein (COMP) as a secreted protein significantly increased in T-PVAT. Recombinant COMP protein significantly inhibited VSMC apoptosis. We conclude that T-PVAT exerts anti-apoptosis effect on VSMCs and attenuates AAA formation, which is possibly attributed to the secretome of browning adipocytes.
Collapse
Affiliation(s)
- Chun-Ling Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu-Na Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Lei Yao
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jun-Ping Li
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zeng-Hui Zhang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Zhao-Qi Huang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Si-Xu Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Yu-Ling Zhang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China
| | - Jing-Feng Wang
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Yang-Xin Chen
- Department of Cardiology, Guangdong Provincial Key Laboratory of Arrhythmia and Electrophysiology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| | - Zhao-Yu Liu
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, 510120, China.
| |
Collapse
|
10
|
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
|
11
|
Wang H, Li J, Wang Z, Tian Y, Li C, Jin F, Li J, Wang L. Perivascular brown adipocytes-derived kynurenic acid relaxes blood vessel via endothelium PI3K-Akt-eNOS pathway. Biomed Pharmacother 2022; 150:113040. [PMID: 35658210 DOI: 10.1016/j.biopha.2022.113040] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 04/15/2022] [Accepted: 04/25/2022] [Indexed: 11/28/2022] Open
Abstract
OBJECTIVE Several metabolites from the kynurenine pathway of tryptophan metabolism play a critical role in vascular function and vascular wall remodeling. This study aimed to test whether metabolite kynurenic acid (KYNA) from the kynurenine pathway relaxes blood vessels. APPROACH AND RESULTS We employed histological staining, in vitro cell culture, Western blotting, real-time PCR, and nitric oxide detection to validate kynurenine aminotransferase (KAT) localization in the vasculature as well as KYNA action on endothelial cells. We also detected vascular reactivity by organ chamber and monitored blood pressure by telemetry to investigate the regulation effect of KYNA on vascular tone. The results presented that perivascular adipose tissue (PVAT) from mice thoracic aorta had robust staining of anti-KAT1 and KYNA than PVAT from the abdominal aorta and mesenteric artery, which is consistent with the expression profile of brown adipocyte marker uncoupling protein 1. KYNA, metabolized from kynurenine by KAT, relaxed pre-contracted both aortic ring and mesenteric artery. In addition, KYNA derived from KAT in PVAT participates in the cross-talk between PVAT and vessel by mediating PVAT inhibition on agonist-induced thoracic aorta contraction. Furthermore, intraperitoneal injection of KYNA in mice reduced blood pressure. The vessel relaxation effect of KYNA was through the endothelium-dependent PI3K-Akt-eNOS pathway. Finally, the high-fat diet decreased KAT1 expression in perithoracic aortic fat and led to KYNA reduction in blood. CONCLUSIONS Our research identified KYNA generated by KAT as a novel perivascular brown adipocyte-derived vascular relaxation factor and suggests that KYNA reduction is a critical event in vascular dysfunction under obese condition.
Collapse
Affiliation(s)
- Huan Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China; Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street SE, Atlanta, GA 30303, USA
| | - Jian Li
- Center for Molecular and Translational Medicine, Georgia State University, 157 Decatur Street SE, Atlanta, GA 30303, USA
| | - Zheng Wang
- Department of Cardiology, The First Affiliated Hospital of Hainan Medical University, Hainan 570102, PR China
| | - Yanfeng Tian
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Chunlei Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| | - Feng Jin
- Foreign Language Teaching Department, Gui Zhou University of Traditional Chinese Medicine, Gui Zhou 550025, PR China
| | - Jia Li
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China.
| | - Lanfeng Wang
- Department of Cardiology, The First Affiliated Hospital of Harbin Medical University, Harbin 150001, PR China
| |
Collapse
|
12
|
Kociszewska K, Deja MA, Malinowski M, Kowalówka A. Vasorelaxing properties of the perivascular tissue of the human radial artery. Eur J Cardiothorac Surg 2022; 61:1423-1429. [PMID: 35134901 PMCID: PMC9728790 DOI: 10.1093/ejcts/ezac074] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 12/07/2021] [Accepted: 01/24/2022] [Indexed: 10/27/2023] Open
Abstract
OBJECTIVES Perivascular adipose tissue (PVAT) surrounding the human internal thoracic artery exhibits anticontractile and vasorelaxing properties associated with the adipocyte-derived relaxing factor (ADRF). The goal of our study was to assess if perivascular tissue of the human radial artery (RA) also exhibits such anticontractile/vasorelaxant properties. It could be especially relevant in preventing RA spasms. METHODS The study was performed on isolated segments of human pedicled RA. Its skeletonized fragments were suspended on stainless steel wire hooks and gradually contracted with serotonin to establish the concentration-effect relationship in the presence/absence of PVAT. Skeletonized arterial segments were precontracted with a single dose of 10-6 M serotonin (EC80). The 5-ml PVAT aliquots (from PVAT incubated in Krebs-Henseleit solution) were transferred to the RA tissue bath resulting in its relaxation. Subsequently, we investigated if ADRF is dependent on endothelial vasorelaxants (nitric oxide and prostacyclin). We attempted to find the potassium channel responsible for mediating the activity of ADRF using different potassium channel blockers. RESULTS RA without PVAT contracted more strongly in response to serotonin compared to RA with PVAT [Emax: 108.3 (20.2) vs 76.1 (13.5) mN]. The PVAT aliquot relaxed precontracted RA rings at 43% (2.4%) [72.2 (15.6) to 41.0 (5.6) mN]. ADRF is independent of endothelial vasorelaxants; hence, the addition of NG-monomethyl-l-arginine and indomethacin did not change the vasorelaxant response. Neither of the potassium channel blockers participated in the activity of ADRF. CONCLUSIONS PVAT of human RA exhibits anticontractile/vasorelaxant properties that are inherently associated with ADRF secretion. We confirmed the endothelial-independent mechanism of the activity of ADRF. However, we failed to find the potassium channel responsible for the action of ADRF.
Collapse
Affiliation(s)
- Karolina Kociszewska
- Department of Cardiac Surgery, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Marek Andrzej Deja
- Department of Cardiac Surgery, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Marcin Malinowski
- Department of Cardiac Surgery, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| | - Adam Kowalówka
- Department of Cardiac Surgery, Medical University of Silesia, School of Medicine in Katowice, Katowice, Poland
| |
Collapse
|
13
|
Patel D, Sharma D, Mandal P. Gut Microbiota: Target for Modulation of Gut-Liver-Adipose Tissue Axis in Ethanol-Induced Liver Disease. Mediators Inflamm 2022; 2022:4230599. [PMID: 35633655 PMCID: PMC9142314 DOI: 10.1155/2022/4230599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/19/2022] [Accepted: 04/20/2022] [Indexed: 02/06/2023] Open
Abstract
Consumption of alcohol (ethanol) in various forms has been an integral part of human civilization. Since ages, it also has been an important cause of death and health impairment across the globe. Ethanol-mediated liver injury, known as alcoholic liver disease (ALD), is caused by surplus intake of alcohol. Several studies have proposed the different pathways that may be lead to ALD. One of the factors that may affect the cytochrome P450 (CYP2E1) metabolic pathway is gut dysbiosis. The gut microbiota produces various compounds that play an important role in regulating healthy functions of distal organs such as the adipose tissue and liver. Dysbiosis causes bacteremia, hepatic encephalopathy, and increased intestinal permeability. Recent clinical studies have found better understanding of the gut and liver axis. Another factor that may affect the ALD pathway is dysfunction of adipose tissue metabolism. Moreover, dysfunction of adipose tissue leads to ectopic fat deposition within the liver and disturbs lipid metabolism by increasing lipolysis/decreasing lipogenesis and impaired glucose tolerance of adipose tissue which leads to ectopic fat deposition within the liver. Adipokine secretion of resistin, leptin, and adiponectin is adversely modified upon prolonged alcohol consumption. In the combination of these two factors, a proinflammatory state is developed within the patient leading to the progression of ALD. Thus, the therapeutic approach for treatments and prevention for liver cirrhosis patients must be focused on the gut-liver-adipose tissue network modification with the use of probiotics, synbiotics, and prebiotics. This review is aimed at the effect of ethanol on gut and adipose tissue in both rodent and human alcoholic models.
Collapse
Affiliation(s)
- Dhara Patel
- PD Patel Institute of Science and Technology, Charotar University of Science and Technology, 388421, Changa, Gujarat, India
| | - Dixa Sharma
- PD Patel Institute of Science and Technology, Charotar University of Science and Technology, 388421, Changa, Gujarat, India
| | - Palash Mandal
- PD Patel Institute of Science and Technology, Charotar University of Science and Technology, 388421, Changa, Gujarat, India
| |
Collapse
|
14
|
Wang Y, Yildiz F, Struve A, Kassmann M, Markó L, Köhler MB, Luft FC, Gollasch M, Tsvetkov D. Aging Affects K V7 Channels and Perivascular Adipose Tissue-Mediated Vascular Tone. Front Physiol 2021; 12:749709. [PMID: 34899382 PMCID: PMC8662361 DOI: 10.3389/fphys.2021.749709] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 10/26/2021] [Indexed: 12/04/2022] Open
Abstract
Aging is an independent risk factor for hypertension, cardiovascular morbidity, and mortality. However, detailed mechanisms linking aging to cardiovascular disease are unclear. We studied the aging effects on the role of perivascular adipose tissue and downstream vasoconstriction targets, voltage-dependent KV7 channels, and their pharmacological modulators (flupirtine, retigabine, QO58, and QO58-lysine) in a murine model. We assessed vascular function of young and old mesenteric arteries in vitro using wire myography and membrane potential measurements with sharp electrodes. We also performed bulk RNA sequencing and quantitative reverse transcription-polymerase chain reaction tests in mesenteric arteries and perivascular adipose tissue to elucidate molecular underpinnings of age-related phenotypes. Results revealed impaired perivascular adipose tissue-mediated control of vascular tone particularly via KV7.3–5 channels with increased age through metabolic and inflammatory processes and release of perivascular adipose tissue-derived relaxation factors. Moreover, QO58 was identified as novel pharmacological vasodilator to activate XE991-sensitive KCNQ channels in old mesenteric arteries. Our data suggest that targeting inflammation and metabolism in perivascular adipose tissue could represent novel approaches to restore vascular function during aging. Furthermore, KV7.3–5 channels represent a promising target in cardiovascular aging.
Collapse
Affiliation(s)
- Yibin Wang
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Fatima Yildiz
- Faculty of Medicine, Istanbul Medeniyet University, Istanbul, Turkey
| | - Andrey Struve
- Department of Ear, Throat and Nose Diseases, I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| | - Mario Kassmann
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Greifswald, Germany
| | - Lajos Markó
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - May-Britt Köhler
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Friedrich C Luft
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Maik Gollasch
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Greifswald, Germany
| | - Dmitry Tsvetkov
- Charité Medical Faculty, Experimental and Clinical Research Center (ECRC), Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany.,Department of Internal Medicine and Geriatrics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
15
|
The Role of Obesity-Induced Perivascular Adipose Tissue (PVAT) Dysfunction in Vascular Homeostasis. Nutrients 2021; 13:nu13113843. [PMID: 34836100 PMCID: PMC8621306 DOI: 10.3390/nu13113843] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/17/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is an additional special type of adipose tissue surrounding blood vessels. Under physiological conditions, PVAT plays a significant role in regulation of vascular tone, intravascular thermoregulation, and vascular smooth muscle cell (VSMC) proliferation. PVAT is responsible for releasing adipocytes-derived relaxing factors (ADRF) and perivascular-derived relaxing factors (PDRF), which have anticontractile properties. Obesity induces increased oxidative stress, an inflammatory state, and hypoxia, which contribute to PVAT dysfunction. The exact mechanism of vascular dysfunction in obesity is still not well clarified; however, there are some pathways such as renin-angiotensin-aldosterone system (RAAS) disorders and PVAT-derived factor dysregulation, which are involved in hypertension and endothelial dysfunction development. Physical activity has a beneficial effect on PVAT function among obese patients by reducing the oxidative stress and inflammatory state. Diet, which is the second most beneficial non-invasive strategy in obesity treatment, may have a positive impact on PVAT-derived factors and may restore the balance in their concentration.
Collapse
|
16
|
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
|
17
|
Barros PR, Costa TJ, Akamine EH, Tostes RC. Vascular Aging in Rodent Models: Contrasting Mechanisms Driving the Female and Male Vascular Senescence. FRONTIERS IN AGING 2021; 2:727604. [PMID: 35821995 PMCID: PMC9261394 DOI: 10.3389/fragi.2021.727604] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Accepted: 08/25/2021] [Indexed: 12/12/2022]
Abstract
Increasing scientific interest has been directed to sex as a biological and decisive factor on several diseases. Several different mechanisms orchestrate vascular function, as well as vascular dysfunction in cardiovascular and metabolic diseases in males and females. Certain vascular sex differences are present throughout life, while others are more evident before the menopause, suggesting two important and correlated drivers: genetic and hormonal factors. With the increasing life expectancy and aging population, studies on aging-related diseases and aging-related physiological changes have steeply grown and, with them, the use of aging animal models. Mouse and rat models of aging, the most studied laboratory animals in aging research, exhibit sex differences in many systems and physiological functions, as well as sex differences in the aging process and aging-associated cardiovascular changes. In the present review, we introduce the most common aging and senescence-accelerated animal models and emphasize that sex is a biological variable that should be considered in aging studies. Sex differences in the cardiovascular system, with a focus on sex differences in aging-associated vascular alterations (endothelial dysfunction, remodeling and oxidative and inflammatory processes) in these animal models are reviewed and discussed.
Collapse
Affiliation(s)
- Paula R. Barros
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Tiago J. Costa
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Eliana H. Akamine
- Department of Pharmacology, Institute of Biomedical Science, University of São Paulo, São Paulo, Brazil
- *Correspondence: Rita C. Tostes, ; Eliana H. Akamine,
| | - Rita C. Tostes
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
- *Correspondence: Rita C. Tostes, ; Eliana H. Akamine,
| |
Collapse
|
18
|
Kumar RK, Kaiser LM, Rockwell CE, Watts SW. Interleukin-10 does not contribute to the anti-contractile nature of PVAT in health. Vascul Pharmacol 2021; 138:106838. [PMID: 33540122 PMCID: PMC8174099 DOI: 10.1016/j.vph.2021.106838] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/23/2020] [Accepted: 01/27/2021] [Indexed: 01/24/2023]
Abstract
Perivascular adipose tissue (PVAT) is protective and reduces contraction of blood vessels in health. PVAT is composed of adipocytes, multiple types of immune cells and stromal cells. Interleukin (IL)-10, an anti-inflammatory cytokine usually produced by T cells, B cells and macrophages, was identified as one of the highly expressed (mRNA) cytokines in the mesenteric PVAT of healthy rats. One report suggested that exogenous IL-10 causes relaxation of mouse mesenteric arteries, also suggesting that IL-10 maybe a potential anti-contractile factor. Hence, we hypothesized that PVAT-derived IL-10 causes vasorelaxation and/or reduces vasoconstriction, thus contributing to the anti-contractile nature of PVAT in health. Mesenteric arteries from rats and mice expressed the receptor for IL-10 (in tunica intima and media) as determined by immunohistochemistry. Mesenteric resistance arteries for rats and superior mesenteric artery for mice were used for isometric contractility studies. Increasing concentrations [0.4-100 ng/mL] of recombinant rat/mouse (rr/mr) IL-10 or vehicle was directly added to half-maximally constricted (phenylephrine, PE) vessels (without PVAT, with endothelium). IL-10 did not cause a direct vasorelaxation. Further, the ability of rrIL-10 to cause a rightward or downward shift of a vasoconstriction-response curve was tested in the rat. The vessels were incubated with rrIL-10 [100 ng/mL or 10 ng/mL] or vehicle for 1.5 h in the tissue bath followed by a cumulative PE [10-8-10-4 M] or U46619 [10-10-10-5 M] response curve. The maximal contractions and EC50 values were similar in IL-10 incubated vessels vs vehicle. Thus, acute exposure of exogenous IL-10 did not reduce local vasoconstriction. To further test if endogenous IL-10 from PVAT was anti-contractile, superior mesenteric arteries from IL-10 WT and KO mice, with and without PVAT, were subjected to increasing concentrations of PE. The anti-contractile nature of PVAT was preserved with both short-term and prolonged depletion (using younger and older mice, respectively) of endogenous IL-10 in males and females. Contrary to our hypothesis, PVAT-derived IL-10 neither caused vasorelaxation nor reduced local vasoconstriction directly/indirectly. Therefore, IL-10 does not contribute to the anti-contractile nature of PVAT in healthy rodents.
Collapse
Affiliation(s)
- R K Kumar
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA.
| | - L M Kaiser
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| | - C E Rockwell
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| | - S W Watts
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| |
Collapse
|
19
|
Liao J, Yin H, Huang J, Hu M. Dysfunction of perivascular adipose tissue in mesenteric artery is restored by aerobic exercise in high-fat diet induced obesity. Clin Exp Pharmacol Physiol 2021; 48:697-703. [PMID: 32893373 DOI: 10.1111/1440-1681.13404] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 08/09/2020] [Accepted: 08/25/2020] [Indexed: 02/06/2023]
Abstract
This study investigated the function of perivascular adipose tissue (PVAT) on vascular contractility within resistant arteries in high-fat diet induced obese rats after long-term aerobic exercise. Male Sprague-Dawley rats were subjected to normal diet control group (N-CTRL), normal diet exercise group (N-EX), high-fat diet control group (H-CTRL), and high-fat diet exercise group (H-EX) (n = 8 in each group). After intervention, adipose tissues morphology was observed. Vasomotor function of mesenteric arteries with or without PVAT were assessed; mesenteric PVAT isolated from each group were transferred to chambers bath with untreated vessels (without PVAT) to evaluate the independent effect. Isolated PVAT was further pre-treated with inhibitor of cystathionine-γ-lyase (CSE), a key hydrogen sulphide (H2 S) enzyme. Results showed that the size of lipid droplet around mesenteric arteries from H-EX was significantly reduced (P < .05); uncoupling protein1 (UCP1) in PVAT from H-EX was enhanced. In N-CTRL, N-EX, and H-EX, vessels without PVAT showed higher sensitivity to serotonin (5-HT) than that with intact PVAT. Vascular tension by 5-HT was significantly reduced in H-EX than H-CTRL (P < .05) in vessels with PVAT. Transferred PVAT from H-EX compared with H-CTRL significantly reduced vascular sensitivity to 5-HT (P < .05), and this effect was eliminated through inhibiting CSE. In summary, the anti-contractile effect of PVAT on resistance artery was impaired in obesity but restored by long-term aerobic exercise. The function of PVAT modified by obesity or by exercise has an independent influence on vascular reactivity, and PVAT derived H2 S may participate in this process.
Collapse
Affiliation(s)
- Jingwen Liao
- Guangdong Provincial Key Laboratory of Sports and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Honggang Yin
- Guangdong Provincial Key Laboratory of Sports and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Junhao Huang
- Guangdong Provincial Key Laboratory of Sports and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
| | - Min Hu
- Guangdong Provincial Key Laboratory of Sports and Health Promotion, Scientific Research Center, Guangzhou Sport University, Guangzhou, China
- Department of Sports and Health, Guangzhou Sport University, Guangzhou, China
| |
Collapse
|
20
|
Dwaib HS, AlZaim I, Eid AH, Obeid O, El-Yazbi AF. Modulatory Effect of Intermittent Fasting on Adipose Tissue Inflammation: Amelioration of Cardiovascular Dysfunction in Early Metabolic Impairment. Front Pharmacol 2021; 12:626313. [PMID: 33897419 PMCID: PMC8062864 DOI: 10.3389/fphar.2021.626313] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022] Open
Abstract
Cardiometabolic syndrome (CMS) is a cluster of maladaptive cardiovascular, renal, thrombotic, inflammatory, and metabolic disorders. It confers a high risk of cardiovascular mortality and morbidity. CMS is triggered by major shifts in lifestyle and dietary habits with increased consumption of refined, calorie-dense diets. Evidence indicates that diet-induced CMS is linked to Adipose tissue (AT) inflammation. This led to the proposal that adipose inflammation may be involved in metabolic derangements, such as insulin resistance and poor glycemic control, as well as the contribution to the inflammatory process predisposing patients to increased cardiovascular risk. Therefore, in the absence of direct pharmacological interventions for the subclinical phase of CMS, time restricted feeding regimens were anticipated to alleviate early metabolic damage and subsequent comorbidities. These regimens, referred to as intermittent fasting (IF), showed a strong positive impact on the metabolic state of obese and non-obese human subjects and animal models, positive AT remodeling in face of overnutrition and high fat diet (HFD) consumption, and improved CV outcomes. Here, we summarize the available evidence on the role of adipose inflammation in triggering cardiovascular impairment in the context of diet induced CMS with an emphasis on the involvement of perivascular adipose tissue. As well, we propose some possible molecular pathways linking intermittent fasting to the ameliorative effect on adipose inflammation and cardiovascular dysfunction under such circumstances. We highlight a number of targets, whose function changes in perivascular adipose tissue inflammation and could be modified by intermittent fasting acting as a novel approach to ameliorate the inflammatory status.
Collapse
Affiliation(s)
- Haneen S. Dwaib
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ibrahim AlZaim
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Biochemistry and Molecular Genetics, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ali H. Eid
- Department of Basic Medical Sciences, College of Medicine, QU Health, Qatar University, Doha, Qatar
- Biomedical and Pharmaceutical Research Unit, QU Health, Qatar University, Doha, Qatar
| | - Omar Obeid
- Department of Nutrition and Food Sciences, Faculty of Agricultural and Food Sciences, American University of Beirut, Beirut, Lebanon
| | - Ahmed F. El-Yazbi
- Department of Pharmacology and Toxicology, Faculty of Medicine, American University of Beirut, Beirut, Lebanon
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
- Faculty of Pharmacy, Al-Alamein International University, Alamein, Egypt
| |
Collapse
|
21
|
Mazzotta C, Basu S, Gower AC, Karki S, Farb MG, Sroczynski E, Zizza E, Sarhan A, Pande AN, Walsh K, Dobrilovic N, Gokce N. Perivascular Adipose Tissue Inflammation in Ischemic Heart Disease. Arterioscler Thromb Vasc Biol 2021; 41:1239-1250. [PMID: 33504180 DOI: 10.1161/atvbaha.120.315865] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
OBJECTIVE There is growing recognition that adipose tissue-derived proatherogenic mediators contribute to obesity-related cardiovascular disease. We sought to characterize regional differences in perivascular adipose tissue (PVAT) phenotype in relation to atherosclerosis susceptibility. Approach and Results: We examined thoracic PVAT samples in 34 subjects (body mass index 32±6 kg/m2, age 59±11 years) undergoing valvular, aortic, or coronary artery bypass graft surgeries and performed transcriptomic characterization using whole-genome expression profiling and quantitative polymerase chain reaction analyses. We identified a highly inflamed region of PVAT surrounding the human aortic root in close proximity to coronary takeoff and adjoining epicardial fat. In subjects undergoing coronary artery bypass graft, we found 300 genes significantly upregulated (false discovery rate Q<0.1) in paired samples of PVAT surrounding the aortic root compared with nonatherosclerotic left internal mammary artery. Genes encoding proteins mechanistically implicated in atherogenesis were enriched in aortic PVAT consisting of signaling pathways linked to inflammation, WNT (wingless-related integration site) signaling, matrix remodeling, coagulation, and angiogenesis. Overexpression of several proatherogenic transcripts, including IL1β, CCL2 (MCP-1), and IL6, were confirmed by quantitative polymerase chain reaction and significantly bolstered in coronary artery disease subjects. Angiographic coronary artery disease burden quantified by the Gensini score positively correlated with the expression of inflammatory genes in PVAT. Moreover, periaortic adipose inflammation was markedly higher in obese subjects with striking upregulation (≈8-fold) of IL1β expression compared to nonobese individuals. CONCLUSIONS Proatherogenic mediators that originate from dysfunctional PVAT may contribute to vascular disease mechanisms in human vessels. Moreover, PVAT may adopt detrimental properties under obese conditions that play a key role in the pathophysiology of ischemic heart disease. Graphic Abstract: A graphic abstract is available for this article.
Collapse
Affiliation(s)
- Celestina Mazzotta
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Sanchita Basu
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Adam C Gower
- Clinical and Translational Science Institute (A.C.G.), Boston University School of Medicine, MA
| | | | - Melissa G Farb
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Emily Sroczynski
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Elaina Zizza
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Anas Sarhan
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Ashvin N Pande
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| | - Kenneth Walsh
- Hematovascular Biology Center and the Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville (K.W.)
| | - Nikola Dobrilovic
- Division of Cardiac Surgery, Department of Surgery, Boston Medical Center, MA (N.D.)
| | - Noyan Gokce
- Evans Department of Medicine and Whitaker Cardiovascular Institute (C.M., S.B., S.K., M.G.F., E.S., E.Z., A.S., A.N.P., N.G.), Boston University School of Medicine, MA
| |
Collapse
|
22
|
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: 49] [Impact Index Per Article: 12.3] [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
|
23
|
Saxton SN, Heagerty AM, Withers SB. Perivascular adipose tissue: An immune cell metropolis. Exp Physiol 2020; 105:1440-1443. [PMID: 32648363 DOI: 10.1113/ep087872] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 07/06/2020] [Indexed: 12/16/2022]
Abstract
NEW FINDINGS What is the topic of this review? The review discusses how eosinophils can contribute to the function of perivascular adipose tissue and explores the mechanisms involved. What advances does it highlight? Understanding the communication between the cell populations that constitute perivascular adipose tissue function is important for exploring therapeutic options in the treatment of obesity-related cardiovascular complications. This article highlights that eosinophils are able to contribute directly to healthy perivascular adipose tissue function. These immune cells contribute to adrenergic signalling and nitric oxide- and adiponectin-dependent mechanisms in perivascular adipose tissue. ABSTRACT Perivascular adipose tissue is a heterogeneous tissue that surrounds most blood vessels in the body. This review focuses on the contribution of eosinophils located within the adipose tissue to vascular contractility. A high-fat diet reduces the number of these immune cells within perivascular adipose tissue, and this loss is linked to an increase in vascular contractility and hypertension. We explored the mechanisms by which eosinophils contribute to this function using genetically modified mice, ex vivo assessment of contractility and pharmacological tools. We found that eosinophils contribute to adrenergic signalling and nitric oxide- and adiponectin-dependent mechanisms in perivascular adipose tissue. It is now important to explore whether manipulation of these pathways in obesity can alleviate cardiovascular complications, in order to determine whether eosinophils are a valid target for obesity-related disease.
Collapse
Affiliation(s)
- S N Saxton
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - A M Heagerty
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - S B Withers
- Division of Cardiovascular Sciences, University of Manchester, Manchester, UK.,School of Science, Engineering and Environment, University of Salford, Salford, UK.,Salford Royal NHS Foundation Trust, Salford Royal Hospitals NHS Foundation Trust, Salford, UK
| |
Collapse
|
24
|
The Effect of Rice Bran Extract on Arterial Blood Pressure, Hepatic Steatosis, and Inflammation in Mice Fed with a High-Fat Diet. J Nutr Metab 2020; 2020:8374287. [PMID: 32685208 PMCID: PMC7341425 DOI: 10.1155/2020/8374287] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Revised: 06/05/2020] [Accepted: 06/09/2020] [Indexed: 12/20/2022] Open
Abstract
Background Inflammation and hypertension are primary mechanisms involving in obesity-associated adverse effects of a high-fat diet. The aim of this study was to evaluate the effects of rice bran extract (RBE) on arterial blood pressure, hepatic steatosis, inflammation, and oxidative stress in high-fat diet (HFD)-induced obese mice. Methods Male ICR mice were divided into four groups, including a normal-diet control group, a high-fat diet (HFD) (60% kcal from fat) group, an HFD group treated with RBE (220 mg/kg/day), and an HFD group treated with 1100 mg/kg/day for eight weeks. Besides body weight and arterial blood pressure, we determined liver values of total cholesterol, triglyceride, as well as percent body fat, tumor necrosis factor-α (TNF-α), malondialdehyde (MDA), nuclear factor kappa-B (NF-κB), matrix metalloprotease-9 (MMP-9), cyclooxygenase-2 (COX-2), and mRNA endothelial nitric oxide synthase (eNOS). Results The HFD group had increased body weight, increased systolic and diastolic blood pressure, liver total cholesterol, triglyceride, NF-κB, COX-2 and MMP-9 protein levels, and decreased mRNA eNOS in the aorta. Mice of the HFD group receiving RBE had reduced diastolic blood pressure, as well as significantly decreased liver and serum TNF-α and MDA levels in the liver, and reduced NF-κB levels in both the liver and heart. Conclusions These results demonstrate that RBE decreases diastolic blood pressure, the liver lipid droplet accumulation, liver and myocardial NF-κB, myocardial COX-2 and MMP-9 protein levels, and oxidative stress. Moreover, RBE may improve endothelial function and may alleviate adverse health effects associated with obesity including obesity-associated hypertension.
Collapse
|
25
|
Targeting perivascular and epicardial adipose tissue inflammation: therapeutic opportunities for cardiovascular disease. Clin Sci (Lond) 2020; 134:827-851. [PMID: 32271386 DOI: 10.1042/cs20190227] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/07/2023]
Abstract
Major shifts in human lifestyle and dietary habits toward sedentary behavior and refined food intake triggered steep increase in the incidence of metabolic disorders including obesity and Type 2 diabetes. Patients with metabolic disease are at a high risk of cardiovascular complications ranging from microvascular dysfunction to cardiometabolic syndromes including heart failure. Despite significant advances in the standards of care for obese and diabetic patients, current therapeutic approaches are not always successful in averting the accompanying cardiovascular deterioration. There is a strong relationship between adipose inflammation seen in metabolic disorders and detrimental changes in cardiovascular structure and function. The particular importance of epicardial and perivascular adipose pools emerged as main modulators of the physiology or pathology of heart and blood vessels. Here, we review the peculiarities of these two fat depots in terms of their origin, function, and pathological changes during metabolic deterioration. We highlight the rationale for pharmacological targeting of the perivascular and epicardial adipose tissue or associated signaling pathways as potential disease modifying approaches in cardiometabolic syndromes.
Collapse
|
26
|
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: 28] [Impact Index Per Article: 5.6] [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
|
27
|
Abstract
Leptin is a hormone that plays a major role as mediator of long-term regulation of energy balance, suppressing food intake, and stimulating weight loss. More recently, important physiological roles other than controlling appetite and energy expenditure have been suggested for leptin, including neuroendocrine, reparative, reproductive, and immune functions. These emerging peripheral roles let hypothesize that leptin can modulate also cancer progression. Indeed, many studies have demonstrated that elevated chronic serum concentrations of leptin, frequently seen in obese subjects, represent a stimulatory signal for tumor growth. Current knowledge indicates that also different non-tumoral cells resident in tumor microenvironment may respond to leptin creating a favorable soil for cancer cells. In addition, leptin is produced also within the tumor microenvironment creating the possibility for paracrine and autocrine action. In this review, we describe the main mechanisms that regulate peripheral leptin availability and how leptin can shape tumor microenvironment.
Collapse
|
28
|
Perivascular Adipose Tissue Modulation of Neurogenic Vasorelaxation of Rat Mesenteric Arteries. J Cardiovasc Pharmacol 2020; 75:21-30. [DOI: 10.1097/fjc.0000000000000761] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
29
|
Ramasubramanian B, Reddy PH. Are TallyHo Mice A True Mouse Model for Type 2 Diabetes and Alzheimer’s Disease? J Alzheimers Dis 2019; 72:S81-S93. [DOI: 10.3233/jad-190613] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
| | - P. Hemachandra Reddy
- Internal Medicine Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Cell Biology & Biochemistry Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Pharmacology & Neuroscience Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
- Neurology Department, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
30
|
Awata WM, Gonzaga NA, Borges VF, Silva CB, Tanus-Santos JE, Cunha FQ, Tirapelli CR. Perivascular adipose tissue contributes to lethal sepsis-induced vasoplegia in rats. Eur J Pharmacol 2019; 863:172706. [DOI: 10.1016/j.ejphar.2019.172706] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022]
|
31
|
Tsvetkov D, Kolpakov E, Kassmann M, Schubert R, Gollasch M. Distinguishing Between Biological and Technical Replicates in Hypertension Research on Isolated Arteries. Front Med (Lausanne) 2019; 6:126. [PMID: 31281816 PMCID: PMC6595250 DOI: 10.3389/fmed.2019.00126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 05/22/2019] [Indexed: 11/27/2022] Open
Abstract
Perivascular adipose tissue (PVAT) is implicated in the pathophysiology of cardiovascular disease, especially in obese individuals in which the quantity of renal and visceral PVAT is markedly increased. The control of arterial tone by PVAT has emerged as a relatively new field of experimental hypertension research. The discovery of this prototype of vasoregulation has been mostly inferred from data obtained using wire myography. Currently, there is a major discussion on distinguishing between biological vs. technical replicates in biomedical studies, which resulted in numerous guidelines being published on planning studies and publishing data by societies, journals, and associations. Experimental study designs are determined depending on how the experimentator distinguishes between biological vs. technical replicates. These definitions determine the ultimate standards required for making submissions to certain journals. In this article, we examine possible outcomes of different experimental study designs on PVAT control of arterial tone using isolated arteries. Based on experimental data, we determine the sample size and power of statistical analyses for such experiments. We discuss whether n-values should correspond to the number of arterial rings and analyze the resulting effects if those numbers are averaged to provide a single N-value per animal, or whether the hierarchical statistical method represents an alternative for analyzing such kind of data. Our analyses show that that the data (logEC50) from (+) PVAT to (-) PVAT arteries are clustered. Intraclass correlation (ICC) was 31.4%. Moreover, it appeared that the hierarchical approach was better than regular statistical tests as the analyses revealed by a better goodness of fit (v2-2LL test). Based on our results, we propose to use at least three independent arterial rings from each from three animals or at least seven arterial rings from each from two animals for each group, i.e., (+) PVAT vs. (-) PVAT. Finally, we discuss a clinical situation where distinguishing between biological vs. technical replicates can lead to absurd situations in clinical decision makings. We conclude that discrimination between biological vs. technical replicates is helpful in experimental studies but is difficult to implement in everyday's clinical practice.
Collapse
Affiliation(s)
- Dmitry Tsvetkov
- Experimental and Clinical Research Center, A Joint Cooperation Between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics and Interfaculty Center of Pharmacogenomics and Drug Research, University of Tübingen, Tübingen, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Evgeniy Kolpakov
- Experimental and Clinical Research Center, A Joint Cooperation Between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Mario Kassmann
- Experimental and Clinical Research Center, A Joint Cooperation Between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim and European Center of Angioscience, Research Division Cardiovascular Physiology, Medical Faculty Mannheim of the University Heidelberg, Mannheim, Germany
- Department of Physiology, Medical Faculty, Augsburg University, Augsburg, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center, A Joint Cooperation Between the Charité Medical Faculty and the Max Delbrück Center for Molecular Medicine (MDC), Berlin, Germany
- Medical Clinic for Nephrology and Internal Intensive Care, Charité University Medicine, Berlin, Germany
| |
Collapse
|
32
|
Gharakhanian R, Su S, Aprahamian T. Vascular Endothelial Growth Factor-A Deficiency in Perivascular Adipose Tissue Impairs Macrovascular Function. Front Physiol 2019; 10:687. [PMID: 31258484 PMCID: PMC6587635 DOI: 10.3389/fphys.2019.00687] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2019] [Accepted: 05/16/2019] [Indexed: 01/10/2023] Open
Abstract
Objective: Thoracic perivascular adipose tissue (PVAT) has been shown to release factors that influence the functioning of neighboring vascular tissue. Cardiovascular complications of obesity are on the rise; therefore, this study set out to determine if adipose-specific ablation of vascular endothelial growth factor-A (VEGF-A) plays a role in the maintenance of aortic structure and function. Methods: Adipose-specific VEGF-A-deficient mice were previously generated. Fabp4cre(+). VEGFflox/flox and Fabp4cre(−). VEGFflox/flox mice were maintained on chow diet. PVAT gene expression was measured with real-time quantitative PCR. Aortic vasomotor response was assessed with isometric tension measurements. Collagen deposition was analyzed histologically in the vascular media and compared using ratiometric pigment density. Results: PVAT-specific adiponectin expression was decreased in Fabp4cre(+). VEGFflox/flox mice. Isometric tension measurements revealed a dose-dependent dysfunction in response to acetylcholine within the distal aortic segment of Fabp4cre(+). VEGFflox/flox. Fabp4cre(+). VEGFflox/flox mice exhibited increased aortic deposition of collagen within the thoracic adventitial and medial spaces. Conclusion: These data demonstrate that decreased expression of VEGF-A within the surrounding adipose tissue microenvironment of the thoracic aorta has a detrimental effect on aortic integrity and vascular function. Modulation of angiogenic pathways within PVAT may offer an important avenue toward the treatment of adipose tissue dysfunction in obesity and its vascular complications.
Collapse
Affiliation(s)
- Raffi Gharakhanian
- Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, United States
| | - Shi Su
- Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, United States
| | - Tamar Aprahamian
- Division of Graduate Medical Sciences, Boston University School of Medicine, Boston, MA, United States.,Renal Section, Department of Medicine, Boston University School of Medicine, Boston, MA, United States.,The Center for Metabolic Health, Boston Medical Center, Boston, MA, United States
| |
Collapse
|
33
|
Nava E, Llorens S. The Local Regulation of Vascular Function: From an Inside-Outside to an Outside-Inside Model. Front Physiol 2019; 10:729. [PMID: 31244683 PMCID: PMC6581701 DOI: 10.3389/fphys.2019.00729] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Accepted: 05/27/2019] [Indexed: 01/22/2023] Open
Abstract
Our understanding of the regulation of vascular function, specifically that of vasomotion, has evolved dramatically over the past few decades. The classic conception of a vascular system solely regulated by circulating hormones and sympathetic innervation gave way to a vision of a local regulation. Initially by the so-called, autacoids like prostacyclin, which represented the first endothelium-derived paracrine regulator of smooth muscle. This was the prelude of the EDRF-nitric oxide age that has occupied vascular scientists for nearly 30 years. Endothelial cells revealed to have the ability to generate numerous mediators besides prostacyclin and nitric oxide (NO). The need to classify these substances led to the coining of the terms: endothelium-derived relaxing, hyperpolarizing and contracting factors, which included various prostaglandins, thromboxane A2, endothelin, as well numerous candidates for the hyperpolarizing factor. The opposite layer of the vascular wall, the adventitia, eventually and for a quite short period of time, enjoyed the attention of some vascular physiologists. Adventitial fibroblasts were recognized as paracrine cells to the smooth muscle because of their ability to produce some substances such as superoxide. Remarkably, this took place before our awareness of the functional potential of another adventitial cell, the adipocyte. Possibly, because the perivascular adipose tissue (PVAT) was systematically removed during the experiments as considered a non-vascular artifact tissue, it took quite long to be considered a major source of paracrine substances. These are now being integrated in the vast pool of mediators synthesized by adipocytes, known as adipokines. They include hormones involved in metabolic regulation, like leptin or adiponectin; classic vascular mediators like NO, angiotensin II or catecholamines; and inflammatory mediators or adipocytokines. The first substance studied was an anti-contractile factor named adipose-derived relaxing factor of uncertain chemical nature but possibly, some of the relaxing mediators mentioned above are behind this factor. This manuscript intends to review the vascular regulation from the point of view of the paracrine control exerted by the cells present in the vascular environment, namely, endothelial, adventitial, adipocyte and vascular stromal cells.
Collapse
Affiliation(s)
- Eduardo Nava
- Department of Medical Sciences, Faculty of Medicine of Albacete, Centro Regional de Investigaciones Biomédicas (CRIB), University of Castilla-La Mancha, Albacete, Spain
| | - Silvia Llorens
- Department of Medical Sciences, Faculty of Medicine of Albacete, Centro Regional de Investigaciones Biomédicas (CRIB), University of Castilla-La Mancha, Albacete, Spain
| |
Collapse
|
34
|
Nóbrega N, Araújo NF, Reis D, Facine LM, Miranda CAS, Mota GC, Aires RD, Capettini LDSA, Cruz JDS, Bonaventura D. Hydrogen peroxide and nitric oxide induce anticontractile effect of perivascular adipose tissue via renin angiotensin system activation. Nitric Oxide 2019; 84:50-59. [PMID: 30611765 DOI: 10.1016/j.niox.2018.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 11/23/2018] [Accepted: 12/31/2018] [Indexed: 01/31/2023]
Abstract
The perivascular adipose tissue (PVAT) is located around the adventitia, composed primarily by adipocytes, stromal cells, leukocytes, fibroblasts and capillaries. It is well described that PVAT is an important modulator of the vascular tone being considered a biologically active tissue, releasing both vasoconstrictor and vasodilators factors. The literature shows that the anti-contractile effect induced by PVAT may be due to activation of the renin-angiotensin system (RAS). AIM Investigate whether the renin-angiotensin system participates in the effect exerted by perivascular adipose tissue on the vascular tone. METHODS AND RESULTS For this study we used thoracic aorta from Balb/c mice and performed vascular reactivity, nitric oxide and hydrogen peroxide quantification using selective probes and fluorescence microscopy, immunofluorescence to locate receptors and enzymes involved in this response. Our results demonstrated that perivascular adipose tissue induces an anti-contractile effect in endothelium-independent manner and involves Mas and AT2 receptors participation with subsequent PI3K/Akt pathway activation. This pathway culminated with nitric oxide and hydrogen peroxide production by neuronal nitric oxide synthase, being hydrogen peroxide most relevant for the anti-contractile effect of perivascular adipose tissue. CONCLUSION For the first time in the literature, our results show the presence of Mas and AT2 receptors, as well as, nitric oxide synthase on perivascular adipose tissue. Furthermore, our results show the involvement of Mas and AT2 receptors and consequently nitric oxide synthase activation in the anti-contractile effect exerted by perivascular adipose tissue.
Collapse
Affiliation(s)
- Natália Nóbrega
- Laboratory of Vascular Pharmacology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Natália Ferreira Araújo
- Laboratory of Vascular Pharmacology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Daniela Reis
- Laboratory of Vascular Pharmacology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Larissa Moreira Facine
- Laboratory of Vascular Pharmacology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Claudiane Aparecida S Miranda
- Laboratory of Vascular Pharmacology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Gianne Campos Mota
- Laboratory of Vascular Biology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Rosária Dias Aires
- Laboratory of Excitatory Membranes, Department of Biochemistry and Immunology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | | | - Jader Dos Santos Cruz
- Laboratory of Excitatory Membranes, Department of Biochemistry and Immunology, Institute of Biological Science, Federal University of Minas Gerais, Brazil
| | - Daniella Bonaventura
- Laboratory of Vascular Pharmacology, Department of Pharmacology, Institute of Biological Science, Federal University of Minas Gerais, Brazil.
| |
Collapse
|
35
|
Gollasch M, Welsh DG, Schubert R. Perivascular adipose tissue and the dynamic regulation of K v 7 and K ir channels: Implications for resistant hypertension. Microcirculation 2018; 25. [PMID: 29211322 DOI: 10.1111/micc.12434] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Accepted: 11/30/2017] [Indexed: 12/20/2022]
Abstract
Resistant hypertension is defined as high blood pressure that remains uncontrolled despite treatment with at least three antihypertensive drugs at adequate doses. Resistant hypertension is an increasingly common clinical problem in older age, obesity, diabetes, sleep apnea, and chronic kidney disease. Although the direct vasodilator minoxidil was introduced in the early 1970s, only recently has this drug been shown to be particularly effective in a subgroup of patients with treatment-resistant or uncontrolled hypertension. This pharmacological approach is interesting from a mechanistic perspective as minoxidil is the only clinically used K+ channel opener today, which targets a subclass of K+ channels, namely KATP channels in VSMCs. Beside KATP channels, two other classes of VSMC K+ channels could represent novel effective targets for treatment of resistant hypertension, namely Kv 7 (KCNQ) and inward rectifier potassium (Kir 2.1) channels. Interestingly, these channels are unique among VSMC potassium channels. First, both have been implicated in the control of microvascular tone by perivascular adipose tissue. Second, they exhibit biophysical properties strongly controlled and regulated by membrane voltage, but not intracellular calcium. This review focuses on Kv 7 (Kv 7.1-5) and Kir (Kir 2.1) channels in VSMCs as potential novel drug targets for treatment of resistant hypertension, particularly in comorbid conditions such as obesity and metabolic syndrome.
Collapse
Affiliation(s)
- Maik Gollasch
- Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Experimental and Clinical Research Center (ECRC) - a joint cooperation between the Charité - University Medicine Berlin and the Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Donald G Welsh
- Department of Physiology and Pharmacology, Western University, London, ON, Canada
| | - Rudolf Schubert
- Centre for Biomedicine and Medical Technology Mannheim (CBTM), Research Division Cardiovascular Physiology, Medical Faculty Mannheim, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
36
|
Lian X, Beer-Hammer S, König GM, Kostenis E, Nürnberg B, Gollasch M. RXFP1 Receptor Activation by Relaxin-2 Induces Vascular Relaxation in Mice via a Gα i2-Protein/PI3Kß/γ/Nitric Oxide-Coupled Pathway. Front Physiol 2018; 9:1234. [PMID: 30233409 PMCID: PMC6131674 DOI: 10.3389/fphys.2018.01234] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Accepted: 08/15/2018] [Indexed: 01/10/2023] Open
Abstract
Background: Relaxins are small peptide hormones, which are novel candidate molecules that play important roles in cardiometablic syndrome. Relaxins are structurally related to the insulin hormone superfamily, which provide vasodilatory effects by activation of G-protein-coupled relaxin receptors (RXFPs) and stimulation of endogenous nitric oxide (NO) generation. Recently, relaxin could be demonstrated to activate Gi proteins and phosphoinositide 3-kinase (PI3K) pathways in cultured endothelial cells in vitro. However, the contribution of the Gi-PI3K pathway and their individual components in relaxin-dependent relaxation of intact arteries remains elusive. Methods: We used Gαi2- (Gnai2-/-) and Gαi3-deficient (Gnai3-/-) mice, pharmacological tools and wire myography to study G-protein-coupled signaling pathways involved in relaxation of mouse isolated mesenteric arteries by relaxins. Human relaxin-1, relaxin-2, and relaxin-3 were tested. Results: Relaxin-2 (∼50% relaxation at 10-11 M) was the most potent vasodilatory relaxin in mouse mesenteric arteries, compared to relaxin-1 and relaxin-3. The vasodilatory effects of relaxin-2 were inhibited by removal of the endothelium or treatment of the vessels with N (G)-nitro-L-arginine methyl ester (L-NAME, endothelial nitric oxide synthase (eNOS) inhibitor) or simazine (RXFP1 inhibitor). The vasodilatory effects of relaxin-2 were absent in arteries of mice treated with pertussis toxin (PTX). They were also absent in arteries isolated from Gnai2-/- mice, but not from Gnai3-/- mice. The effects were not affected by FR900359 (Gαq protein inhibitor) or PI-103 (PI3Kα inhibitor), but inhibited by TGX-221 (PI3Kβ inhibitor) or AS-252424 (PI3Kγ inhibitor). Simazine did not influence the anti-contractile effect of perivascular adipose tissue. Conclusion: Our data indicate that relaxin-2 produces endothelium- and NO-dependent relaxation of mouse mesenteric arteries by activation of RXFP1 coupled to Gi2-PI3K-eNOS pathway. Targeting vasodilatory Gi-protein-coupled RXFP1 pathways may provide promising opportunities for drug discovery in endothelial dysfunction and cardiometabolic disease.
Collapse
Affiliation(s)
- Xiaoming Lian
- Experimental and Clinical Research Center (ECRC), Charité - University Medicine Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sandra Beer-Hammer
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen, Germany
| | - Gabriele M König
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Evi Kostenis
- Institute for Pharmaceutical Biology, University of Bonn, Bonn, Germany
| | - Bernd Nürnberg
- Department of Pharmacology and Experimental Therapy, Institute of Experimental and Clinical Pharmacology and Toxicology, Eberhard Karls University Hospitals and Clinics, and Interfaculty Center of Pharmacogenomics and Drug Research (ICePhA), Tübingen, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center (ECRC), Charité - University Medicine Berlin and Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany.,Medical Clinic for Nephrology and Internal Intensive Care, Charité Campus Virchow Klinikum, Berlin, Germany
| |
Collapse
|
37
|
Kumar RK, Darios ES, Burnett R, Thompson JM, Watts SW. Fenfluramine-induced PVAT-dependent contraction depends on norepinephrine and not serotonin. Pharmacol Res 2018; 140:43-49. [PMID: 30189295 DOI: 10.1016/j.phrs.2018.08.024] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/24/2018] [Accepted: 08/27/2018] [Indexed: 12/24/2022]
Abstract
Perivascular adipose tissue (PVAT) modulates vascular tone and altered PVAT function is observed in vascular diseases such as hypertension and atherosclerosis. We discovered that the PVAT surrounding rat thoracic aorta (RA) and the superior mesenteric artery (SMA) contain significant amounts of 5-hydroxytryptamine (5-HT). We hypothesized that the 5-HT contained within the PVAT is functional and vasoactive. Isolated tissue baths were used for isometric contractility studies and high performance liquid chromatography was used to quantitatively measure amines in the PVAT and release studies. The 5-HT releaser fenfluramine (10 nM-100 μM) was tested for its ability to contract arteries with and without PVAT. Contraction was reported as a percentage of the initial contraction to 10 μM phenylephrine. The RA with PVAT contracted to fenfluramine to a greater maximum (98 ± 10%) than RA without PVAT (24 ± 4%), while no difference in contraction of SMA to maximum fenfluramine with (78 ± 2%) and without (75 ± 6%) PVAT was observed. Contradicting our hypothesis, the maximum contraction of RA with PVAT to fenfluramine was diminished by the alpha-1 adrenoreceptor antagonist prazosin (100 nM; vehicle: 71 ± 4%, prazosin: 24 ± 2%) and the norepinephrine transporter (NET) inhibitor nisoxetine (1 μM; vehicle: 71 ± 4%, nisoxetine: 25 ± 4%) but not the 5-HT2A/2C receptor antagonist ketanserin (10 nM) or serotonin specific reuptake inhibitor fluoxetine (10 μM). To test if fenfluramine caused release of 5-HT or NE from PVAT, PVAT from RA was incubated with vehicle or fenfluramine (10 μM-10 mM), and amines released into the incubating buffer were quantified. A pronounced concentration-dependent NE-release (more than 5-HT) was observed. Collectively, this research illustrates the pharmacology of fenfluramine to primarily stimulate NE release (better than 5-HT) in a NET-dependent manner, leading to vasoconstriction. This adds additional support to PVAT as being an important reservoir of amines.
Collapse
Affiliation(s)
- Ramya K Kumar
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA.
| | - Emma S Darios
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| | - Robert Burnett
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| | - Janice M Thompson
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| | - Stephanie W Watts
- Department of Pharmacology and Toxicology, Michigan State University, MI, USA
| |
Collapse
|
38
|
Gonzaga NA, Awata WMC, do Vale GT, Marchi KC, Muniz JJ, Tanus-Santos JE, Tirapelli CR. Perivascular adipose tissue protects against the vascular dysfunction induced by acute ethanol intake: Role of hydrogen peroxide. Vascul Pharmacol 2018; 111:44-53. [PMID: 30157459 DOI: 10.1016/j.vph.2018.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 07/04/2018] [Accepted: 08/25/2018] [Indexed: 01/04/2023]
Abstract
AIM We investigated the consequences of acute ethanol intake on the anti-contractile effect of perivascular adipose tissue (PVAT). METHODS The effects of a single dose of ethanol (1 g/kg; p.o. gavage) on the vascular function were assessed within 30 min in male Wistar rats. RESULTS Ethanol decreased the relaxation induced by acetylcholine and increased the contraction induced by phenylephrine in endothelium-intact, but not in endothelium-denuded aortas without PVAT. The vascular dysfunction induced by ethanol was not observed in aortic rings with PVAT. Nω-Nitro-l-arginine methyl ester (L-NAME), NG-nitro-l-arginine (L-NNA) and 1H-(1,2,4)oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), but not tiron or tempol, increased the contraction induced by phenylephrine in endothelium-intact aortas with PVAT from control and ethanol-treated rats. Catalase increased phenylephrine-induced contraction in aortas with PVAT from ethanol-treated rats, but not from control rats. Conversely, inhibition of catalase with aminotriazole decreased phenylephrine-induced contraction in aortas from ethanol-treated rats. Treatment with ethanol increased hydrogen peroxide (H2O2) levels and decreased catalase activity in aortas with PVAT. Ethanol increased superoxide anion (O2-) generation in aortas with or without PVAT. Superoxide dismutase (SOD) activity was not affected by ethanol intake. In situ quantification of H2O2 using 2'7'dichlorodihydrofluorescein diacetate (DCFH-DA) revealed increased levels of H2O2 in periaortic PVAT from ethanol-treated rats. However, in situ evaluation of nitric oxide (NO) in both aorta and PVAT showed no differences between groups. CONCLUSIONS Our study provides novel evidence that the periaortic PVAT protects against the vascular dysfunction induced by acute ethanol intake through a mechanism that involves increased generation of H2O2.
Collapse
Affiliation(s)
- Natália A Gonzaga
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Laboratório de Farmacologia, DEPCH, Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Wanessa M C Awata
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Laboratório de Farmacologia, DEPCH, Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Gabriel T do Vale
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Laboratório de Farmacologia, DEPCH, Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Katia C Marchi
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil; Laboratório de Farmacologia, DEPCH, Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Jaqueline J Muniz
- Laboratório de Farmacologia, DEPCH, Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil
| | - Jose E Tanus-Santos
- Programa de Pós-Graduação em Farmacologia, Faculdade de Medicina de Ribeirão Preto, Universidade de São Paulo (USP), Ribeirão Preto, SP, Brazil
| | - Carlos R Tirapelli
- Laboratório de Farmacologia, DEPCH, Escola de Enfermagem de Ribeirão Preto, USP, Ribeirão Preto, SP, Brazil.
| |
Collapse
|
39
|
Zemančíková A, Török J. Effect of perivascular adipose tissue on arterial adrenergic contractions in normotensive and hypertensive rats with high fructose intake. Physiol Res 2018; 66:S537-S544. [PMID: 29355382 DOI: 10.33549/physiolres.933798] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The aim of this study was to investigate the effect of high fructose intake associated with moderate increase in adiposity on rat arterial adrenergic responses and their modulation by perivascular adipose tissue (PVAT). After eight-week-lasting substitution of drinking water with 10 % fructose solution in adult normotensive Wistar-Kyoto rats (WKY) and spontaneously hypertensive rats (SHR), their systolic blood pressure, plasma triglycerides, and relative liver weight were elevated when compared to their respective control groups. Moreover, in SHR, body weight and relative heart weight were increased after treatment with fructose. In superior mesenteric arteries, PVAT exerted inhibitory influence on adrenergic contractile responses and this effect was markedly stronger in control WKY than in SHR. In fructose-administered WKY, arterial adrenergic contractions were substantially reduced in comparison with the control group; this was caused mainly by enhancement of anticontractile action of PVAT. The diminution of the mesenteric arterial contractions was not observed after fructose treatment in SHR. We conclude that the increase in body adiposity due to fructose overfeeding in rats might have prehypertensive effect. However, in WKY it might cause PVAT-dependent and independent reduction in arterial contractile responses to adrenergic stimuli, which could attenuate the pathological elevation in vascular tone.
Collapse
Affiliation(s)
- A Zemančíková
- Institute of Normal and Pathological Physiology, Slovak Academy of Sciences, Bratislava, Slovak Republic.
| | | |
Collapse
|
40
|
Bussey CE, Withers SB, Saxton SN, Bodagh N, Aldous RG, Heagerty AM. β 3 -Adrenoceptor stimulation of perivascular adipocytes leads to increased fat cell-derived NO and vascular relaxation in small arteries. Br J Pharmacol 2018; 175:3685-3698. [PMID: 29980164 PMCID: PMC6109217 DOI: 10.1111/bph.14433] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 05/04/2018] [Accepted: 06/19/2018] [Indexed: 01/13/2023] Open
Abstract
Background and Purpose In response to noradrenaline, healthy perivascular adipose tissue (PVAT) exerts an anticontractile effect on adjacent small arterial tissue. Organ bath solution transfer experiments have demonstrated the release of PVAT‐derived relaxing factors that mediate this function. The present studies were designed to investigate the mechanism responsible for the noradrenaline‐induced PVAT anticontractile effect. Experimental Approach In vitro rat small arterial contractile function was assessed using wire myography in the presence and absence of PVAT and the effects of sympathomimetic stimulation on the PVAT environment explored using Western blotting and assays of organ bath buffer. Key Results PVAT elicited an anticontractile effect in response to noradrenaline but not phenylephrine stimulation. In arteries surrounded by intact PVAT, the β3‐adrenoceptor agonist, CL‐316243, reduced the vasoconstrictor effect of phenylephrine but not noradrenaline. Kv7 channel inhibition using XE 991 reversed the noradrenaline‐induced anticontractile effect in exogenously applied PVAT studies. Adrenergic stimulation of PVAT with noradrenaline and CL‐316243, but not phenylephrine, was associated with increased adipocyte‐derived NO production, and the contractile response to noradrenaline was augmented following incubation of exogenous PVAT with L‐NMMA. PVAT from eNOS−/− mice had no anticontractile effect. Assays of adipocyte cAMP demonstrated an increase with noradrenaline stimulation implicating Gαs signalling in this process. Conclusions and Implications We have shown that adipocyte‐located β3‐adrenoceptor stimulation leads to activation of Gαs signalling pathways with increased cAMP and the release of adipocyte‐derived NO. This process is dependent upon Kv7 channel function. We conclude that adipocyte‐derived NO plays a central role in anticontractile activity when rodent PVAT is stimulated by noradrenaline.
Collapse
Affiliation(s)
- Charlotte E Bussey
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Sarah B Withers
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK.,Environment and Life Sciences, University of Salford, Salford, UK
| | - Sophie N Saxton
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Neil Bodagh
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Robert G Aldous
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| | - Anthony M Heagerty
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
41
|
Yang L, Chen L, Lu X, Tan A, Chen Y, Li Y, Peng X, Yuan S, Cai D, Yu Y. Peri-ovarian adipose tissue contributes to intraovarian control during folliculogenesis in mice. Reproduction 2018; 156:133-144. [DOI: 10.1530/rep-18-0120] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 05/29/2018] [Indexed: 12/30/2022]
Abstract
Peri-ovarian adipose tissue (POAT) is a kind of intra-abdominal white adipose tissue that is present surrounding the ovaries in rodents. Recent studies demonstrated that POAT-deficient mice displayed a phenotype of delayed antral follicular development, for which decreases in serum estrogen, serum FSH and FSHR levels were responsible. However, folliculogenesis is regulated by endocrine signals and also modulated by a number of locally produced intraovarian factors whose acts are both autocrine and paracrine. Here, we used a model of surgical removal of POAT unilaterally and contralateral ovaries as controls, as both were under the same endocrine control, to assess the paracrine effect of the POAT on folliculogenesis. Surgical removal of unilateral POAT resulted in delayed antral follicular development and the increased number of atretic follicles, accompanied by decreased levels of intraovarian adipokines and growth factors, lipid accumulation and steroidogenic enzyme expression. POAT-deficient ovaries displayed compensatory increased expressions of intraovarian genes, such as Vegf and Adpn for angiogenesis, Acc, Fasn, and Gapdh involved in lipogenesis and Fshr in response to FSH stimulation. Furthermore, we demonstrated that removal of POAT promoted follicular apoptosis, caused retention of cytoplasmic YAP and inhibited PTEN-AKT-mTOR activation. These alterations were observed only in the POAT-deficient ovaries but not in the contralateral ovaries (with POAT), which suggests that a paracrine interaction between POAT and ovaries is important for normal folliculogenesis.
Collapse
|
42
|
Intrauterine exposure to metformin: Evaluation of endothelial and perivascular adipose tissue function in abdominal aorta of adult offspring. Life Sci 2018; 207:72-79. [PMID: 29852188 DOI: 10.1016/j.lfs.2018.05.050] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 05/19/2018] [Accepted: 05/28/2018] [Indexed: 01/08/2023]
Abstract
The biguanide metformin (MET) has been used during pregnancy for treatment of polycystic ovary syndrome and gestational diabetes. MET crosses the placenta and maternal treatment can expose the progeny to this drug during important phases of body development. Direct vascular protective effects have been described with the treatment of metformin. Nevertheless, it is unclear whether intrauterine exposure to metformin is safe for the vascular system of offspring. Thus, the present study aimed to investigate the intrinsic effects of metformin exposure in utero in the offspring abdominal aorta reactivity, in the presence and absence of perivascular adipose tissue (PVAT) and endothelium. For this, Wistar rats were treated with metformin 293 mg/kg/day (MET) or water (CTR) by gavage during the gestational period. The abdominal aorta reactivity to phenylephrine, acetylcholine, and sodium nitroprusside was evaluated in male adult offspring. It was observed that abdominal aorta relaxation was similar between MET and CTR groups in the presence or absence of PVAT. In addition, the contraction to phenylephrine was similar between MET and CTR groups in the presence and absence of PVAT and endothelium. Therefore, metformin exposure during pregnancy had no intrinsic effect on the offspring abdominal aorta PVAT and endothelial function, demonstrating it to be safe to the vascular system of the offspring.
Collapse
|
43
|
Wang N, Kuczmanski A, Dubrovska G, Gollasch M. Palmitic Acid Methyl Ester and Its Relation to Control of Tone of Human Visceral Arteries and Rat Aortas by Perivascular Adipose Tissue. Front Physiol 2018; 9:583. [PMID: 29875688 PMCID: PMC5974537 DOI: 10.3389/fphys.2018.00583] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 05/01/2018] [Indexed: 01/01/2023] Open
Abstract
Background: Perivascular adipose tissue (PVAT) exerts anti-contractile effects on visceral arteries by release of various perivascular relaxing factors (PVRFs) and opening voltage-gated K+ (Kv) channels in vascular smooth muscle cells (VSMCs). Palmitic acid methyl ester (PAME) has been proposed as transferable PVRF in rat aorta. Here, we studied PVAT regulation of arterial tone of human mesenteric arteries and clarified the contribution of Kv channels and PAME in the effects. Methods: Wire myography was used to measure vasocontractions of mesenteric artery rings from patients undergoing abdominal surgery. Isolated aortic rings from Sprague-Dawley rats were studied for comparison. PVAT was either left intact or removed from the arterial rings. Vasocontractions were induced by external high K+ (60 mM), serotonin (5-HT) or phenylephrine. PAME (10 nM−3 μM) was used as vasodilator. Kv channels were blocked by XE991, a Kv7 (KCNQ) channel inhibitor, or by 4-aminopyridine, a non-specific Kv channel inhibitor. PAME was measured in bathing solutions incubated with rat peri-aortic or human visceral adipose tissue. Results: We found that PVAT displayed anti-contractile effects in both human mesenteric arteries and rat aortas. The anti-contractile effects were inhibited by XE991 (30 μM). PAME (EC50 ~1.4 μM) was capable to produce relaxations of PVAT-removed rat aortas. These effects were abolished by XE991 (30 μM), but not 4-aminopyridine (2 mM) or NDGA (10 μM), a lipoxygenases inhibitor. The cytochrome P450 epoxygenase inhibitor 17-octadecynoic acid (ODYA 10 μM) and the soluble epoxide hydrolase inhibitor 12-(3-adamantan-1-ylureido)-dodecanoic acid (AUDA 10 μM) slightly decreased PAME relaxations. PAME up to 10 μM failed to induce relaxations of PVAT-removed human mesenteric arteries. 5-HT induced endogenous PAME release from rat peri-aortic adipose tissue, but not from human visceral adipose tissue. Conclusions: Our data also suggest that Kv7 channels are involved in the anti-contractile effects of PVAT on arterial tone in both rat aorta and human mesenteric arteries. PAME could contribute to PVAT relaxations by activating Kv7 channels in rat aorta, but not in human mesenteric arteries.
Collapse
Affiliation(s)
- Ning Wang
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Artur Kuczmanski
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,HELIOS Klinikum Berlin-Buch, Berlin, Germany
| | - Galyna Dubrovska
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Maik Gollasch
- Experimental and Clinical Research Center, Charité - Universitätsmedizin Berlin and Max-Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.,Medical Clinic of Nephrology and Internal Intensive Care, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
44
|
Agabiti-Rosei C, Paini A, De Ciuceis C, Withers S, Greenstein A, Heagerty AM, Rizzoni D. Modulation of Vascular Reactivity by Perivascular Adipose Tissue (PVAT). Curr Hypertens Rep 2018; 20:44. [PMID: 29736674 DOI: 10.1007/s11906-018-0835-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
PURPOSE OF REVIEW In this review, we discuss the role of perivascular adipose tissue (PVAT) in the modulation of vascular contractility and arterial pressure, focusing on the role of the renin-angiotensin-aldosterone system and oxidative stress/inflammation. RECENT FINDINGS PVAT possesses a relevant endocrine-paracrine activity, which may be altered in several pathophysiological and clinical conditions. During the last two decades, it has been shown that PVAT may modulate vascular reactivity. It has also been previously demonstrated that inflammation in adipose tissue may be implicated in vascular dysfunction. In particular, adipocytes secrete a number of adipokines with various functions, as well as several vasoactive factors, together with components of the renin-angiotensin system which may act at local or at systemic level. It has been shown that the anti-contractile effect of PVAT is lost in obesity, probably as a consequence of the development of adipocyte hypertrophy, inflammation, and oxidative stress. Adipose tissue dysfunction is interrelated with inflammation and oxidative stress, thus contributing to endothelial dysfunction observed in several pathological and clinical conditions such as obesity and hypertension. Decreased local adiponectin level, macrophage recruitment and infiltration, and activation of renin-angiotensin-aldosterone system could play an important role in this regard.
Collapse
Affiliation(s)
- Claudia Agabiti-Rosei
- Department of Medicine, Manchester University, Manchester, UK. .,Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy.
| | - Anna Paini
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Carolina De Ciuceis
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| | - Sarah Withers
- Department of Medicine, Manchester University, Manchester, UK
| | - Adam Greenstein
- Department of Medicine, Manchester University, Manchester, UK
| | | | - Damiano Rizzoni
- Clinica Medica, Department of Medical and Surgical Sciences, University of Brescia, c/o 2a Medicina Spedali Civili di Brescia, Piazza Spedali Civili 1, 25100, Brescia, Italy
| |
Collapse
|
45
|
Turaihi AH, Bakker W, van Hinsbergh VWM, Serné EH, Smulders YM, Niessen HWM, Eringa EC. Insulin Receptor Substrate 2 Controls Insulin-Mediated Vasoreactivity and Perivascular Adipose Tissue Function in Muscle. Front Physiol 2018; 9:245. [PMID: 29628894 PMCID: PMC5876319 DOI: 10.3389/fphys.2018.00245] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 03/06/2018] [Indexed: 11/16/2022] Open
Abstract
Introduction: Insulin signaling in adipose tissue has been shown to regulate insulin's effects in muscle. In muscle, perivascular adipose tissue (PVAT) and vascular insulin signaling regulate muscle perfusion. Insulin receptor substrate (IRS) 2 has been shown to control adipose tissue function and glucose metabolism, and here we tested the hypothesis that IRS2 mediates insulin's actions on the vessel wall as well as the vasoactive properties of PVAT. Methods: We studied PVAT and muscle resistance arteries (RA) from littermate IRS2+/+ and IRS2−/− mice and vasoreactivity by pressure myography, vascular insulin signaling, adipokine expression, and release and PVAT morphology. As insulin induced constriction of IRS2+/+ RA in our mouse model, we also exposed RA's of C57/Bl6 mice to PVAT from IRS2+/+ and IRS2−/− littermates to evaluate vasodilator properties of PVAT. Results: IRS2−/− RA exhibited normal vasomotor function, yet a decreased maximal diameter compared to IRS2+/+ RA. IRS2+/+ vessels unexpectedly constricted endothelin-dependently in response to insulin, and this effect was absent in IRS2−/− RA due to reduced ERK1/2activation. For evaluation of PVAT function, we also used C57/Bl6 vessels with a neutral basal effect of insulin. In these experiments insulin (10.0 nM) increased diameter in the presence of IRS2+/+ PVAT (17 ± 4.8, p = 0.014), yet induced a 10 ± 7.6% decrease in diameter in the presence of IRS2−/− PVAT. Adipocytes in IRS2−/− PVAT (1314 ± 161 μm2) were larger (p = 0.0013) than of IRS2+/+ PVAT (915 ± 63 μm2). Adiponectin, IL-6, PAI-1 secretion were similar between IRS2+/+ and IRS2−/− PVAT, as were expression of pro-inflammatory genes (TNF-α, CCL2) and adipokines (adiponectin, leptin, endothelin-1). Insulin-induced AKT phosphorylation in RA was similar in the presence of IRS2−/− and IRS2+/+ PVAT. Conclusion: In muscle, IRS2 regulates both insulin's vasoconstrictor effects, mediating ERK1/2-ET-1 activation, and its vasodilator effects, by mediating the vasodilator effect of PVAT. The regulatory role of IRS2 in PVAT is independent from adiponectin secretion.
Collapse
Affiliation(s)
- Alexander H Turaihi
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| | - Wineke Bakker
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| | - Victor W M van Hinsbergh
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| | - Erik H Serné
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| | - Yvo M Smulders
- Department of Internal Medicine, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| | - Hans W M Niessen
- Department of Pathology and Cardiac Surgery, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| | - Etto C Eringa
- Department of Physiology, Amsterdam Cardiovascular Sciences, VU University Medical Center, Amsterdam, Netherlands
| |
Collapse
|
46
|
Boedtkjer E. Acid-base regulation and sensing: Accelerators and brakes in metabolic regulation of cerebrovascular tone. J Cereb Blood Flow Metab 2018; 38:588-602. [PMID: 28984162 PMCID: PMC5888856 DOI: 10.1177/0271678x17733868] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2017] [Revised: 08/10/2017] [Accepted: 09/06/2017] [Indexed: 12/29/2022]
Abstract
Metabolic regulation of cerebrovascular tone directs blood flow to areas of increased neuronal activity and during disease states partially compensates for insufficient perfusion by enhancing blood flow in collateral blood vessels. Acid-base disturbances frequently occur as result of enhanced metabolism or insufficient blood supply, but despite definitive evidence that acid-base disturbances alter arterial tone, effects of individual acid-base equivalents and the underlying signaling mechanisms are still being debated. H+ is an important intra- and extracellular messenger that modifies cerebrovascular tone. In addition, low extracellular [HCO3-] promotes cerebrovascular contraction through an endothelium-dependent mechanism. CO2 alters arterial tone development via changes in intra- and extracellular pH but it is still controversial whether CO2 also has direct vasomotor effects. Vasocontractile responses to low extracellular [HCO3-] and acute CO2-induced decreases in intracellular pH can counteract H+-mediated vasorelaxation during metabolic and respiratory acidosis, respectively, and may thereby reduce the risk of capillary damage and cerebral edema that could be consequences of unopposed vasodilation. In this review, the signaling mechanisms for acid-base equivalents in cerebral arteries and the mechanisms of intracellular pH control in the arterial wall are discussed in the context of metabolic regulation of cerebrovascular tone and local perfusion.
Collapse
Affiliation(s)
- Ebbe Boedtkjer
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
47
|
Costa RM, Neves KB, Tostes RC, Lobato NS. Perivascular Adipose Tissue as a Relevant Fat Depot for Cardiovascular Risk in Obesity. Front Physiol 2018; 9:253. [PMID: 29618983 PMCID: PMC5871983 DOI: 10.3389/fphys.2018.00253] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 03/06/2018] [Indexed: 12/18/2022] Open
Abstract
Obesity is associated with increased risk of premature death, morbidity, and mortality from several cardiovascular diseases (CVDs), including stroke, coronary heart disease (CHD), myocardial infarction, and congestive heart failure. However, this is not a straightforward relationship. Although several studies have substantiated that obesity confers an independent and additive risk of all-cause and cardiovascular death, there is significant variability in these associations, with some lean individuals developing diseases and others remaining healthy despite severe obesity, the so-called metabolically healthy obese. Part of this variability has been attributed to the heterogeneity in both the distribution of body fat and the intrinsic properties of adipose tissue depots, including developmental origin, adipogenic and proliferative capacity, glucose and lipid metabolism, hormonal control, thermogenic ability, and vascularization. In obesity, these depot-specific differences translate into specific fat distribution patterns, which are closely associated with differential cardiometabolic risks. The adventitial fat layer, also known as perivascular adipose tissue (PVAT), is of major importance. Similar to the visceral adipose tissue, PVAT has a pathophysiological role in CVDs. PVAT influences vascular homeostasis by releasing numerous vasoactive factors, cytokines, and adipokines, which can readily target the underlying smooth muscle cell layers, regulating the vascular tone, distribution of blood flow, as well as angiogenesis, inflammatory processes, and redox status. In this review, we summarize the current knowledge and discuss the role of PVAT within the scope of adipose tissue as a major contributing factor to obesity-associated cardiovascular risk. Relevant clinical studies documenting the relationship between PVAT dysfunction and CVD with a focus on potential mechanisms by which PVAT contributes to obesity-related CVDs are pointed out.
Collapse
Affiliation(s)
- Rafael M Costa
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Karla B Neves
- Institute of Cardiovascular and Medical Sciences, British Heart Foundation, Glasgow Cardiovascular Research Centre, University of Glasgow, Glasgow, United Kingdom
| | - Rita C Tostes
- Department of Pharmacology, Ribeirao Preto Medical School, University of São Paulo, Ribeirao Preto, Brazil
| | - Núbia S Lobato
- Institute of Health Sciences, Federal University of Goias, Jatai, Brazil
| |
Collapse
|
48
|
Almabrouk TAM, White AD, Ugusman AB, Skiba DS, Katwan OJ, Alganga H, Guzik TJ, Touyz RM, Salt IP, Kennedy S. High Fat Diet Attenuates the Anticontractile Activity of Aortic PVAT via a Mechanism Involving AMPK and Reduced Adiponectin Secretion. Front Physiol 2018; 9:51. [PMID: 29479319 PMCID: PMC5812172 DOI: 10.3389/fphys.2018.00051] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2017] [Accepted: 01/16/2018] [Indexed: 01/31/2023] Open
Abstract
Background and aim: Perivascular adipose tissue (PVAT) positively regulates vascular function through production of factors such as adiponectin but this effect is attenuated in obesity. The enzyme AMP-activated protein kinase (AMPK) is present in PVAT and is implicated in mediating the vascular effects of adiponectin. In this study, we investigated the effect of an obesogenic high fat diet (HFD) on aortic PVAT and whether any changes involved AMPK. Methods: Wild type Sv129 (WT) and AMPKα1 knockout (KO) mice aged 8 weeks were fed normal diet (ND) or HFD (42% kcal fat) for 12 weeks. Adiponectin production by PVAT was assessed by ELISA and AMPK expression studied using immunoblotting. Macrophages in PVAT were identified using immunohistochemistry and markers of M1 and M2 macrophage subtypes evaluated using real time-qPCR. Vascular responses were measured in endothelium-denuded aortic rings with or without attached PVAT. Carotid wire injury was performed and PVAT inflammation studied 7 days later. Key results: Aortic PVAT from KO and WT mice was morphologically indistinct but KO PVAT had more infiltrating macrophages. HFD caused an increased infiltration of macrophages in WT mice with increased expression of the M1 macrophage markers Nos2 and Il1b and the M2 marker Chil3. In WT mice, HFD reduced the anticontractile effect of PVAT as well as reducing adiponectin secretion and AMPK phosphorylation. PVAT from KO mice on ND had significantly reduced adiponectin secretion and no anticontractile effect and feeding HFD did not alter this. Wire injury induced macrophage infiltration of PVAT but did not cause further infiltration in KO mice. Conclusions: High-fat diet causes an inflammatory infiltrate, reduced AMPK phosphorylation and attenuates the anticontractile effect of murine aortic PVAT. Mice lacking AMPKα1 phenocopy many of the changes in wild-type aortic PVAT after HFD, suggesting that AMPK may protect the vessel against deleterious changes in response to HFD.
Collapse
Affiliation(s)
- Tarek A M Almabrouk
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Medical School, University of Zawia, Zawia, Libya
| | - Anna D White
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Azizah B Ugusman
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Department of Physiology, National University of Malaysia Medical Centre, Kuala Lumpur, Malaysia
| | - Dominik S Skiba
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Jagiellonian University College of Medicine, Krakow, Poland
| | - Omar J Katwan
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Department of Biochemistry, College of Medicine, University of Diyala, Baqubah, Iraq
| | - Husam Alganga
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Medical School, University of Zawia, Zawia, Libya
| | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.,Jagiellonian University College of Medicine, Krakow, Poland
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Ian P Salt
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Simon Kennedy
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
49
|
Mathew John C, Khaddaj Mallat R, George G, Kim T, Mishra RC, Braun AP. Pharmacologic targeting of endothelial Ca 2+-activated K + channels: A strategy to improve cardiovascular function. Channels (Austin) 2018; 12:126-136. [PMID: 29577810 PMCID: PMC5972810 DOI: 10.1080/19336950.2018.1454814] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Accepted: 03/15/2018] [Indexed: 12/17/2022] Open
Abstract
Endothelial small and intermediate-conductance, Ca2+-activated K+ channels (KCa2.3 and KCa3.1, respectively) play an important role in the regulation of vascular function and systemic blood pressure. Growing evidence indicates that they are intimately involved in agonist-evoked vasodilation of small resistance arteries throughout the circulation. Small molecule activators of KCa2.x and 3.1 channels, such as SKA-31, can acutely inhibit myogenic tone in isolated resistance arteries, induce effective vasodilation in intact vascular beds, such as the coronary circulation, and acutely decrease systemic blood pressure in vivo. The blood pressure-lowering effect of SKA-31, and early indications of improvement in endothelial dysfunction suggest that endothelial KCa channel activators could eventually be developed into a new class of endothelial targeted agents to combat hypertension or atherosclerosis. This review summarises recent insights into the activation of endothelial Ca2+ activated K+ channels in various vascular beds, and how tools, such as SKA-31, may be beneficial in disease-related conditions.
Collapse
Affiliation(s)
- Cini Mathew John
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Rayan Khaddaj Mallat
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Grace George
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Taeyeob Kim
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Ramesh C. Mishra
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Andrew P. Braun
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
50
|
Araujo HN, Victório JA, Valgas da Silva CP, Sponton ACS, Vettorazzi JF, de Moraes C, Davel AP, Zanesco A, Delbin MA. Anti-contractile effects of perivascular adipose tissue in thoracic aorta from rats fed a high-fat diet: role of aerobic exercise training. Clin Exp Pharmacol Physiol 2017; 45:293-302. [PMID: 29265399 DOI: 10.1111/1440-1681.12882] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 10/11/2017] [Accepted: 10/13/2017] [Indexed: 01/19/2023]
Affiliation(s)
- Hygor N. Araujo
- Department of Structural and Functional Biology; Institute of Biology; University of Campinas (UNICAMP); Campinas SP Brazil
| | - Jamaira A. Victório
- Department of Structural and Functional Biology; Institute of Biology; University of Campinas (UNICAMP); Campinas SP Brazil
| | - Carmem P. Valgas da Silva
- Department of Physical Education; Institute of Biosciences; São Paulo State University (UNESP); Rio Claro SP Brazil
| | - Amanda C. S. Sponton
- Department of Structural and Functional Biology; Institute of Biology; University of Campinas (UNICAMP); Campinas SP Brazil
| | - Jean F. Vettorazzi
- Obesity and Comorbidities Research Center; Department of Structural and Functional Biology; Institute of Biology; University of Campinas (UNICAMP); Campinas SP Brazil
| | - Camila de Moraes
- School of Physical Education and Sport of Ribeirão Preto; University of São Paulo (USP); RibeirãoPreto SP Brazil
| | - Ana P. Davel
- Department of Structural and Functional Biology; Institute of Biology; University of Campinas (UNICAMP); Campinas SP Brazil
| | | | - Maria A. Delbin
- Department of Structural and Functional Biology; Institute of Biology; University of Campinas (UNICAMP); Campinas SP Brazil
| |
Collapse
|