1
|
Carlström M, Weitzberg E, Lundberg JO. Nitric Oxide Signaling and Regulation in the Cardiovascular System: Recent Advances. Pharmacol Rev 2024; 76:1038-1062. [PMID: 38866562 DOI: 10.1124/pharmrev.124.001060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 04/30/2024] [Accepted: 05/29/2024] [Indexed: 06/14/2024] Open
Abstract
Nitric oxide (NO) from endothelial NO synthase importantly contributes to vascular homeostasis. Reduced NO production or increased scavenging during disease conditions with oxidative stress contribute to endothelial dysfunction and NO deficiency. In addition to the classical enzymatic NO synthases (NOS) system, NO can also be generated via the nitrate-nitrite-NO pathway. Dietary and pharmacological approaches aimed at increasing NO bioactivity, especially in the cardiovascular system, have been the focus of much research since the discovery of this small gaseous signaling molecule. Despite wide appreciation of the biological role of NOS/NO signaling, questions still remain about the chemical nature of NOS-derived bioactivity. Recent studies show that NO-like bioactivity can be efficiently transduced by mobile NO-ferroheme species, which can transfer between proteins, partition into a hydrophobic phase, and directly activate the soluble guanylyl cyclase-cGMP-protein kinase G pathway without intermediacy of free NO. Moreover, interaction between red blood cells and the endothelium in the regulation of vascular NO homeostasis have gained much attention, especially in conditions with cardiometabolic disease. In this review we discuss both classical and nonclassical pathways for NO generation in the cardiovascular system and how these can be modulated for therapeutic purposes. SIGNIFICANCE STATEMENT: After four decades of intensive research, questions persist about the transduction and control of nitric oxide (NO) synthase bioactivity. Here we discuss NO signaling in cardiovascular health and disease, highlighting new findings, such as the important role of red blood cells in cardiovascular NO homeostasis. Nonclassical signaling modes, like the nitrate-nitrite-NO pathway, and therapeutic opportunities related to the NO system are discussed. Existing and potential pharmacological treatments/strategies, as well as dietary components influencing NO generation and signaling are covered.
Collapse
Affiliation(s)
- Mattias Carlström
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| | - Eddie Weitzberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| | - Jon O Lundberg
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (M.C., E.W., J.O.L.); and Department of Perioperative Medicine and Intensive Care, Karolinska University Hospital, Stockholm, Sweden (E.W.)
| |
Collapse
|
2
|
Zhou M, Hanschmann EM, Römer A, Linn T, Petry SF. The significance of glutaredoxins for diabetes mellitus and its complications. Redox Biol 2024; 71:103043. [PMID: 38377787 PMCID: PMC10891345 DOI: 10.1016/j.redox.2024.103043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 01/13/2024] [Indexed: 02/22/2024] Open
Abstract
Diabetes mellitus is a non-communicable metabolic disease hallmarked by chronic hyperglycemia caused by beta-cell failure. Diabetic complications affect the vasculature and result in macro- and microangiopathies, which account for a significantly increased morbidity and mortality. The rising incidence and prevalence of diabetes is a major global health burden. There are no feasible strategies for beta-cell preservation available in daily clinical practice. Therefore, patients rely on antidiabetic drugs or the application of exogenous insulin. Glutaredoxins (Grxs) are ubiquitously expressed and highly conserved members of the thioredoxin family of proteins. They have specific functions in redox-mediated signal transduction, iron homeostasis and biosynthesis of iron-sulfur (FeS) proteins, and the regulation of cell proliferation, survival, and function. The involvement of Grxs in chronic diseases has been a topic of research for several decades, suggesting them as therapeutic targets. Little is known about their role in diabetes and its complications. Therefore, this review summarizes the available literature on the significance of Grxs in diabetes and its complications. In conclusion, Grxs are differentially expressed in the endocrine pancreas and in tissues affected by diabetic complications, such as the heart, the kidneys, the eye, and the vasculature. They are involved in several pathways essential for insulin signaling, metabolic inflammation, glucose and fatty acid uptake and processing, cell survival, and iron and mitochondrial metabolism. Most studies describe significant changes in glutaredoxin expression and/or activity in response to the diabetic metabolism. In general, mitigated levels of Grxs are associated with oxidative distress, cell damage, and even cell death. The induced overexpression is considered a potential part of the cellular stress-response, counteracting oxidative distress and exerting beneficial impact on cell function such as insulin secretion, cytokine expression, and enzyme activity.
Collapse
Affiliation(s)
- Mengmeng Zhou
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Eva-Maria Hanschmann
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Axel Römer
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Sebastian Friedrich Petry
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
3
|
Evans CJF, Glastras SJ, Tang O, Figtree GA. Therapeutic Potential for Beta-3 Adrenoreceptor Agonists in Peripheral Arterial Disease and Diabetic Foot Ulcers. Biomedicines 2023; 11:3187. [PMID: 38137408 PMCID: PMC10740412 DOI: 10.3390/biomedicines11123187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 11/27/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Annually, peripheral arterial disease is estimated to cost over USD 21 billion and diabetic foot disease an estimated at USD 9-13 billion. Mirabegron is a TGA-approved beta-3 adrenoreceptor agonist, shown to be safe and effective in the treatment of overactive bladder syndrome by stimulating bladder smooth muscle relaxation. In this review, we discuss the potential use of beta-3 adrenoreceptor agonists as therapeutic agents repurposed for peripheral arterial disease and diabetic foot ulcers. The development of both conditions is underpinned by the upregulation of oxidative stress pathways and consequential inflammation and hypoxia. In oxidative stress, there is an imbalance of reactive oxygen species and nitric oxide. Endothelial nitric oxide synthase becomes uncoupled in disease states, producing superoxide and worsening oxidative stress. Agonist stimulation of the beta-3 adrenoreceptor recouples and activates endothelial nitric oxide synthase, increasing the production of nitric oxide. This reduces circulating reactive oxygen species, thus decreasing redox modification and dysregulation of cellular proteins, causing downstream smooth muscle relaxation, improved endothelial function and increased angiogenesis. These mechanisms lead to endothelial repair in peripheral arterial disease and an enhanced perfusion in hypoxic tissue, which will likely improve the healing of chronic ulcers.
Collapse
Affiliation(s)
- Cameron J. F. Evans
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (S.J.G.); (O.T.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Sarah J. Glastras
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (S.J.G.); (O.T.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Diabetes, Endocrinology & Metabolism, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| | - Owen Tang
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (S.J.G.); (O.T.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
| | - Gemma A. Figtree
- Kolling Institute, University of Sydney, Sydney, NSW 2006, Australia; (S.J.G.); (O.T.)
- Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- Department of Cardiology, Royal North Shore Hospital, Northern Sydney Local Health District, Sydney, NSW 2065, Australia
| |
Collapse
|
4
|
Guerra-Ojeda S, Jorda A, Aldasoro C, Vila JM, Valles SL, Arias-Mutis OJ, Aldasoro M. Improvement of Vascular Insulin Sensitivity by Ranolazine. Int J Mol Sci 2023; 24:13532. [PMID: 37686345 PMCID: PMC10487645 DOI: 10.3390/ijms241713532] [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: 07/31/2023] [Revised: 08/22/2023] [Accepted: 08/28/2023] [Indexed: 09/10/2023] Open
Abstract
Ranolazine (RN) is a drug used in the treatment of chronic coronary ischemia. Different clinical trials have shown that RN behaves as an anti-diabetic drug by lowering blood glucose and glycosylated hemoglobin (HbA1c) levels. However, RN has not been shown to improve insulin (IN) sensitivity. Our study investigates the possible facilitating effects of RN on the actions of IN in the rabbit aorta. IN induced vasodilation of the abdominal aorta in a concentration-dependent manner, and this dilatory effect was due to the phosphorylation of endothelial nitric oxide synthase (eNOS) and the formation of nitric oxide (NO). On the other hand, IN facilitated the vasodilator effects of acetylcholine but not the vasodilation induced by sodium nitroprusside. RN facilitated all the vasodilatory effects of IN. In addition, IN decreased the vasoconstrictor effects of adrenergic nerve stimulation and exogenous noradrenaline. Both effects were in turn facilitated by RN. The joint effect of RN with IN induced a significant increase in the ratio of p-eNOS/eNOS and pAKT/AKT. In conclusion, RN facilitated the vasodilator effects of IN, both direct and induced, on the adrenergic system. Therefore, RN increases vascular sensitivity to IN, thus decreasing tissue resistance to the hormone, a key mechanism in the development of type II diabetes.
Collapse
Affiliation(s)
- Sol Guerra-Ojeda
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
| | - Adrian Jorda
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
- Department of Nursing and Podiatry, University of Valencia, 46010 València, Spain
| | - Constanza Aldasoro
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
| | - Jose M. Vila
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
| | - Soraya L. Valles
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
| | - Oscar J Arias-Mutis
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
| | - Martin Aldasoro
- Department of Physiology, University of Valencia, 46010 València, Spain; (S.G.-O.); (A.J.); (C.A.); (J.M.V.); (S.L.V.); (O.J.A.-M.)
| |
Collapse
|
5
|
Balligand JL, Michel LYM. Clinical pharmacology of β-3 adrenergic receptor agonists for cardiovascular diseases. Expert Rev Clin Pharmacol 2023; 16:1073-1084. [PMID: 37728503 DOI: 10.1080/17512433.2023.2193681] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/17/2023] [Indexed: 09/21/2023]
Abstract
INTRODUCTION Few agonists of the third isotype of beta-adrenergic receptors, the β3-adrenoreceptor, are currently used clinically, and new agonists are under development for the treatment of overactive bladder disease. As the receptor is expressed in human cardiac and vascular tissues, it is important to understand their beneficial (or adverse) effect(s) on these targets. AREAS COVERED We discuss the most recent results of clinical trials testing the benefit and safety of β3-adrenoreceptor activation on cardiovascular outcomes in light of current knowledge on the receptor biology, genetic polymorphisms, and agonist pharmacology. EXPERT OPINION While evidence from small clinical trials is limited so far, the β3-agonist, mirabegron seems to be safe in patients at high cardiovascular risk but produces benefits on selected cardiovascular outcomes only at higher than standard doses. Activation of cardiovascular β3-adrenoreceptors deserves to be tested with more potent agonists, such as vibegron.
Collapse
Affiliation(s)
- Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics, Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics, Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| |
Collapse
|
6
|
Bravo FI, Calvo E, López-Villalba RA, Torres-Fuentes C, Muguerza B, García-Ruiz A, Morales D. Valorization of Chicken Slaughterhouse Byproducts to Obtain Antihypertensive Peptides. Nutrients 2023; 15:457. [PMID: 36678328 PMCID: PMC9864718 DOI: 10.3390/nu15020457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 01/09/2023] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
Hypertension (HTN) is the leading cause of premature deaths worldwide and the main preventable risk factor for cardiovascular diseases. Therefore, there is a current need for new therapeutics to manage this condition. In this regard, protein hydrolysates containing antihypertensive bioactive peptides are of increasing interest. Thus, agri-food industry byproducts have emerged as a valuable source to obtain these hydrolysates as they are rich in proteins and inexpensive. Among these, byproducts from animal origin stand out as they are abundantly generated worldwide. Hence, this review is focused on evaluating the potential role of chicken slaughterhouse byproducts as a source of peptides for managing HTN. Several of these byproducts such as blood, bones, skins, and especially, chicken feet have been used to obtain protein hydrolysates with angiotensin-converting enzyme (ACE)-inhibitory activity and blood pressure-lowering effects. An increase in levels of endogenous antioxidant compounds, a reduction in ACE activity, and an improvement of HTN-associated endothelial dysfunction were the mechanisms underlying their effects. However, most of these studies were carried out in animal models, and further clinical studies are needed in order to confirm these antihypertensive properties. This would increase the value of these byproducts, contributing to the circular economy model of slaughterhouses.
Collapse
Affiliation(s)
| | | | | | | | | | - Almudena García-Ruiz
- Nutrigenomics Research Group, Department of Biochemistry and Biotechnology, Universitat Rovira i Virgili, 43007 Tarragona, Spain
| | | |
Collapse
|
7
|
Karimi Galougahi K, Zhang Y, Kienzle V, Liu C, Quek L, Patel S, Lau E, Cordina R, Figtree GA, Celermajer DS. β3 adrenergic agonism: A novel pathway which improves right ventricular-pulmonary arterial hemodynamics in pulmonary arterial hypertension. Physiol Rep 2023; 11:e15549. [PMID: 36597221 PMCID: PMC9810839 DOI: 10.14814/phy2.15549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 06/17/2023] Open
Abstract
Efficacy of therapies that target the downstream nitric oxide (NO) pathway in pulmonary arterial hypertension (PAH) depends on the bioavailability of NO. Reduced NO level in PAH is secondary to "uncoupling" of endothelial nitric oxide synthase (eNOS). Stimulation of β3 adrenergic receptors (β3 ARs) may lead to the recoupling of NOS and therefore be beneficial in PAH. We aimed to examine the efficacy of β3 AR agonism as a novel pathway in experimental PAH. In hypoxia (5 weeks) and Sugen hypoxia (hypoxia for 5 weeks + SU5416 injection) models of PAH, we examined the effects of the selective β3 AR agonist CL316243. We measured echocardiographic indices and invasive right ventricular (RV)-pulmonary arterial (PA) hemodynamics and compared CL316243 with riociguat and sildenafil. We assessed treatment effects on RV-PA remodeling, oxidative stress, and eNOS glutathionylation, an oxidative modification that uncouples eNOS. Compared with normoxic mice, RV systolic pressure was increased in the control hypoxic mice (p < 0.0001) and Sugen hypoxic mice (p < 0.0001). CL316243 reduced RV systolic pressure, to a similar degree to riociguat and sildenafil, in both hypoxia (p < 0.0001) and Sugen hypoxia models (p < 0.03). CL316243 reversed pulmonary vascular remodeling, decreased RV afterload, improved RV-PA coupling efficiency and reduced RV stiffness, hypertrophy, and fibrosis. Although all treatments decreased oxidative stress, CL316243 significantly reduced eNOS glutathionylation. β3 AR stimulation improved RV hemodynamics and led to beneficial RV-PA remodeling in experimental models of PAH. β3 AR agonists may be effective therapies in PAH.
Collapse
Affiliation(s)
- Keyvan Karimi Galougahi
- Heart Research InstituteSydneyAustralia
- Royal Prince Alfred HospitalSydneyAustralia
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | | | | | - Chia‐Chi Liu
- Heart Research InstituteSydneyAustralia
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
- Kolling Institute for Medical ResearchSydneyAustralia
| | - Lake‐Ee Quek
- Charles Perkins CenterUniversity of SydneySydneyAustralia
| | - Sanjay Patel
- Heart Research InstituteSydneyAustralia
- Royal Prince Alfred HospitalSydneyAustralia
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Edmund Lau
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
- Department of Respiratory MedicineRoyal Prince Alfred HospitalSydneyAustralia
| | - Rachael L. Cordina
- Royal Prince Alfred HospitalSydneyAustralia
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| | - Gemma A. Figtree
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
- Kolling Institute for Medical ResearchSydneyAustralia
- Department of CardiologyRoyal North Shore HospitalSydneyAustralia
| | - David S. Celermajer
- Heart Research InstituteSydneyAustralia
- Royal Prince Alfred HospitalSydneyAustralia
- Sydney Medical SchoolFaculty of Medicine and HealthUniversity of SydneySydneyAustralia
| |
Collapse
|
8
|
Kavurma MM, Bursill C, Stanley CP, Passam F, Cartland SP, Patel S, Loa J, Figtree GA, Golledge J, Aitken S, Robinson DA. Endothelial cell dysfunction: Implications for the pathogenesis of peripheral artery disease. Front Cardiovasc Med 2022; 9:1054576. [PMID: 36465438 PMCID: PMC9709122 DOI: 10.3389/fcvm.2022.1054576] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Accepted: 10/24/2022] [Indexed: 08/27/2023] Open
Abstract
Peripheral artery disease (PAD) is caused by occluded or narrowed arteries that reduce blood flow to the lower limbs. The treatment focuses on lifestyle changes, management of modifiable risk factors and vascular surgery. In this review we focus on how Endothelial Cell (EC) dysfunction contributes to PAD pathophysiology and describe the largely untapped potential of correcting endothelial dysfunction. Moreover, we describe current treatments and clinical trials which improve EC dysfunction and offer insights into where future research efforts could be made. Endothelial dysfunction could represent a target for PAD therapy.
Collapse
Affiliation(s)
- Mary M. Kavurma
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Christina Bursill
- South Australian Health and Medical Research Institute, Adelaide, SA, Australia
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA, Australia
| | | | - Freda Passam
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
- Central Clinical School, Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Siân P. Cartland
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
| | - Sanjay Patel
- Heart Research Institute, The University of Sydney, Sydney, NSW, Australia
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Jacky Loa
- Royal Prince Alfred Hospital, Sydney, NSW, Australia
| | - Gemma A. Figtree
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Kolling Institute of Medical Research, Royal North Shore Hospital, Sydney, NSW, Australia
| | - Jonathan Golledge
- Queensland Research Centre for Peripheral Vascular Disease, College of Medicine and Dentistry, James Cook University, Townsville, QLD, Australia
- The Department of Vascular and Endovascular Surgery, Townsville University Hospital, Townsville, QLD, Australia
| | - Sarah Aitken
- Faculty of Health and Medicine, The University of Sydney, Sydney, NSW, Australia
- Concord Institute of Academic Surgery, Concord Hospital, Sydney, NSW, Australia
| | | |
Collapse
|
9
|
Mirabegron relaxes arteries from human visceral adipose tissue through antagonism of α1-adrenergic receptors. Vascul Pharmacol 2022; 146:107094. [PMID: 35934296 DOI: 10.1016/j.vph.2022.107094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 07/13/2022] [Accepted: 08/02/2022] [Indexed: 11/21/2022]
|
10
|
Kajikawa M, Higashi Y. Obesity and Endothelial Function. Biomedicines 2022; 10:biomedicines10071745. [PMID: 35885049 PMCID: PMC9313026 DOI: 10.3390/biomedicines10071745] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/16/2022] [Accepted: 07/16/2022] [Indexed: 02/08/2023] Open
Abstract
Obesity is a major public health problem and is related to increasing rates of cardiovascular morbidity and mortality. Over 1.9 billion adults are overweight or obese worldwide and the prevalence of obesity is increasing. Obesity influences endothelial function through obesity-related complications such as hypertension, dyslipidemia, diabetes, metabolic syndrome, and obstructive sleep apnea syndrome. The excess fat accumulation in obesity causes adipocyte dysfunction and induces oxidative stress, insulin resistance, and inflammation leading to endothelial dysfunction. Several anthropometric indices and imaging modalities that are used to evaluate obesity have demonstrated an association between obesity and endothelial function. In the past few decades, there has been great focus on the mechanisms underlying endothelial dysfunction caused by obesity for the prevention and treatment of cardiovascular events. This review focuses on pathophysiological mechanisms of obesity-induced endothelial dysfunction and therapeutic targets of obesity.
Collapse
Affiliation(s)
- Masato Kajikawa
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan;
| | - Yukihito Higashi
- Division of Regeneration and Medicine, Medical Center for Translational and Clinical Research, Hiroshima University Hospital, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan;
- Department of Regenerative Medicine, Division of Radiation Medical Science, Research Institute for Radiation Biology and Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima 734-8551, Japan
- Correspondence: ; Tel.: +81-82-257-5831
| |
Collapse
|
11
|
Soliman GF, Abdel-Maksoud OM, Khalifa MM, Rashed LA, Ibrahim W, Morsi H, Abdallah H, Bastawy N. Effect of nebivolol on altered skeletal and cardiac muscles induced by dyslipidemia in rats: impact on oxidative and inflammatory machineries. Arch Physiol Biochem 2022; 128:463-473. [PMID: 31876193 DOI: 10.1080/13813455.2019.1693599] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AIM High cholesterol diet is greatly linked to deleterious health consequences. In this work we tried to explore direct effects of high cholesterol diet on striated (skeletal and cardiac) muscle tissues and the mechanisms by which nebivolol could improve such harmful effects. METHODS The study included 24 healthy adult male albino rats weighing 200-220 grams that were assigned into four groups: control group, control drug group, high cholesterol diet fed groups; one untreated the other was treated with nebivolol. RESULTS In the cholesterol fed group, we found decreased blood HDL and NO with elevated total cholesterol, triglycerides, myoglobin, CK, LDH, ALP, in addition to elevated muscle tissue levels of HIF-1, NF-kB, MDA, and decreased expression of both eNOS, reduced GSH. Wire hanging test time was shorter in the high cholesterol group than control group rats, which was confirmed histologically by increased striated muscle fibre thickness and cytochrome area %. Nebivolol treatment ameliorated the effects of high cholesterol diet. CONCLUSION High cholesterol diet caused myopathic changes in rat striated muscle tissues mostly due to oxidative stress associated with enhanced NF-kB and HIF-1 expression. Nebivolol appears beneficial in the management of hypercholesterolaemia-induced striated muscle injury.
Collapse
Affiliation(s)
| | | | | | | | - Walaa Ibrahim
- Department of Medical Biochemistry, Cairo University, Cairo, Egypt
| | - Heba Morsi
- Department of Medical Biochemistry, Cairo University, Cairo, Egypt
| | - Hanan Abdallah
- Department of Medical Histology, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Nermeen Bastawy
- Department of Medical Physiology, Cairo University, Cairo, Egypt
| |
Collapse
|
12
|
Koep JL, Taylor CE, Coombes JS, Bond B, Ainslie PN, Bailey TG. Autonomic control of cerebral blood flow: fundamental comparisons between peripheral and cerebrovascular circulations in humans. J Physiol 2021; 600:15-39. [PMID: 34842285 DOI: 10.1113/jp281058] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 11/25/2021] [Indexed: 01/12/2023] Open
Abstract
Understanding the contribution of the autonomic nervous system to cerebral blood flow (CBF) control is challenging, and interpretations are unclear. The identification of calcium channels and adrenoreceptors within cerebral vessels has led to common misconceptions that the function of these receptors and actions mirror those of the peripheral vasculature. This review outlines the fundamental differences and complex actions of cerebral autonomic activation compared with the peripheral circulation. Anatomical differences, including the closed nature of the cerebrovasculature, and differential adrenoreceptor subtypes, density, distribution and sensitivity, provide evidence that measures on peripheral sympathetic nerve activity cannot be extrapolated to the cerebrovasculature. Cerebral sympathetic nerve activity seems to act opposingly to the peripheral circulation, mediated at least in part by changes in intracranial pressure and cerebral blood volume. Additionally, heterogeneity in cerebral adrenoreceptor distribution highlights region-specific autonomic regulation of CBF. Compensatory chemo- and autoregulatory responses throughout the cerebral circulation, and interactions with parasympathetic nerve activity are unique features to the cerebral circulation. This crosstalk between sympathetic and parasympathetic reflexes acts to ensure adequate perfusion of CBF to rising and falling perfusion pressures, optimizing delivery of oxygen and nutrients to the brain, while attempting to maintain blood volume and intracranial pressure. Herein, we highlight the distinct similarities and differences between autonomic control of cerebral and peripheral blood flow, and the regional specificity of sympathetic and parasympathetic regulation within the cerebrovasculature. Future research directions are outlined with the goal to further our understanding of autonomic control of CBF in humans.
Collapse
Affiliation(s)
- Jodie L Koep
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia.,Children's Health and Exercise Research Centre, Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Chloe E Taylor
- School of Health Sciences, Western Sydney University, Sydney, Australia
| | - Jeff S Coombes
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia
| | - Bert Bond
- Children's Health and Exercise Research Centre, Sport and Health Sciences, College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | - Philip N Ainslie
- Centre for Heart, Lung and Vascular Health, School of Health and Exercise Sciences, University of British Columbia - Okanagan, Kelowna, British Columbia, Canada
| | - Tom G Bailey
- Physiology and Ultrasound Laboratory in Science and Exercise, Centre for Research on Exercise, Physical Activity and Health, School of Human Movement and Nutrition Sciences, The University of Queensland, Brisbane, Queensland, Australia.,School of Nursing, Midwifery and Social Work, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
13
|
Bubb KJ, Harmer JA, Finemore M, Aitken SJ, Ali ZS, Billot L, Chow C, Golledge J, Mister R, Gray MP, Grieve SM, Hamburg N, Keech AC, Patel S, Puttaswamy V, Figtree GA. Protocol for the Stimulating β 3-Adrenergic Receptors for Peripheral Artery Disease (STAR-PAD) trial: a double-blinded, randomised, placebo-controlled study evaluating the effects of mirabegron on functional performance in patients with peripheral arterial disease. BMJ Open 2021; 11:e049858. [PMID: 34588252 PMCID: PMC8479946 DOI: 10.1136/bmjopen-2021-049858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
INTRODUCTION There is currently only one approved medication effective at improving walking distance in people with intermittent claudication. Preclinical data suggest that the β3-adrenergic receptor agonist (mirabegron) could be repurposed to treat intermittent claudication associated with peripheral artery disease. The aim of the Stimulating β3-Adrenergic Receptors for Peripheral Artery Disease (STAR-PAD) trial is to test whether mirabegron improves walking distance in people with intermittent claudication. METHODS AND ANALYSIS The STAR-PAD trial is a Phase II, multicentre, double-blind, randomised, placebo-controlled trial of mirabegron versus placebo on walking distance in patients with PAD. A total of 120 patients aged ≥40 years with stable PAD and intermittent claudication will be randomly assigned (1:1 ratio) to receive either mirabegron (50 mg orally once a day) or matched placebo, for 12 weeks. The primary endpoint is change in peak walking distance as assessed by a graded treadmill test. Secondary endpoints will include: (i) initial claudication distance; (ii) average daily step count and total step count and (iii) functional status and quality of life assessment. Mechanistic substudies will examine potential effects of mirabegron on vascular function, including brachial artery flow-mediate dilatation; MRI assessment of lower limb blood flow, tissue perfusion and arterial stiffness and numbers and angiogenesis potential of endothelial progenitor cells. Given that mirabegron is safe and clinically available for alternative purposes, a positive study is positioned to immediately impact patient care. ETHICS AND DISSEMINATION The STAR-PAD trial is approved by the Northern Sydney Local Health District Human Research Ethics Committee (HREC/18/HAWKE/50). The study results will be published in peer-reviewed medical or scientific journals and presented at scientific meetings, regardless of the study outcomes. TRIAL REGISTRATION NUMBER ACTRN12619000423112; Results.
Collapse
Affiliation(s)
- Kristen J Bubb
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Jason A Harmer
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Meghan Finemore
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Sarah Joy Aitken
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Concord Repatriation General Hospital, Concord, New South Wales, Australia
| | - Zara S Ali
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Laurent Billot
- The George Institute for Global Health, UNSW Sydney, Newtown, New South Wales, Australia
| | - Clara Chow
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- The George Institute for Global Health, UNSW Sydney, Newtown, New South Wales, Australia
- Westmead Applied Research Centre, Faculty of Medicine and Health, University of Sydney, Westmead, New South Wales, Australia
- Department of Cardiology, Westmead Hospital, Westmead, New South Wales, Australia
| | - Jonathan Golledge
- Vascular Biology Unit, James Cook University Queensland Research Centre for Peripheral Vascular Disease, Townsville, Queensland, Australia
| | - Rebecca Mister
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Michael P Gray
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
| | - Stuart M Grieve
- The Heart Research Institute, Newtown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | | | - Anthony C Keech
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- NHMRC Clinical Trials Centre, University of Sydney, Camperdown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
| | - Sanjay Patel
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- The Heart Research Institute, Newtown, New South Wales, Australia
- Charles Perkins Centre, University of Sydney, Camperdown, New South Wales, Australia
- Department of Cardiology, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
| | - Vikram Puttaswamy
- Vascular Surgery, North Shore Private Hospital, Sydney, New South Wales, Australia
| | - Gemma A Figtree
- Faculty of Medicine and Health, University of Sydney, Sydney, New South Wales, Australia
- Kolling Institute of Medical Research, Cardiothoracic and Vascular Health, University of Sydney, St Leonards, New South Wales, Australia
- Department of Cardiology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| |
Collapse
|
14
|
Proniewski B, Bar A, Kieronska-Rudek A, Suraj-Prażmowska J, Buczek E, Czamara K, Majka Z, Czyzynska-Cichon I, Kwiatkowski G, Matyjaszczyk-Gwarda K, Chlopicki S. Systemic Administration of Insulin Receptor Antagonist Results in Endothelial and Perivascular Adipose Tissue Dysfunction in Mice. Cells 2021; 10:cells10061448. [PMID: 34207844 PMCID: PMC8230211 DOI: 10.3390/cells10061448] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 01/07/2023] Open
Abstract
Hyperglycemia linked to diabetes results in endothelial dysfunction. In the present work, we comprehensively characterized effects of short-term hyperglycemia induced by administration of an insulin receptor antagonist, the S961 peptide, on endothelium and perivascular adipose tissue (PVAT) in mice. Endothelial function of the thoracic and abdominal aorta in 12-week-old male C57Bl/6Jrj mice treated for two weeks with S961 infusion via osmotic pumps was assessed in vivo using magnetic resonance imaging and ex vivo by detection of nitric oxide (NO) production using electron paramagnetic resonance spectroscopy. Additional methods were used to analyze PVAT, aortic segments and endothelial-specific plasma biomarkers. Systemic disruption of insulin signaling resulted in severe impairment of NO-dependent endothelial function and a loss of vasoprotective function of PVAT affecting the thoracic as well as abdominal parts of the aorta, however a fall in adiponectin expression and decreased uncoupling protein 1-positive area were more pronounced in the thoracic aorta. Results suggest that dysfunctional PVAT contributes to vascular pathology induced by altered insulin signaling in diabetes, in the absence of fat overload and obesity.
Collapse
Affiliation(s)
- Bartosz Proniewski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Anna Bar
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Anna Kieronska-Rudek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
- Faculty of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531 Krakow, Poland
| | - Joanna Suraj-Prażmowska
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Elżbieta Buczek
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Krzysztof Czamara
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Zuzanna Majka
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
- Faculty of Chemistry, Jagiellonian University, Gronostajowa 2, 30-387 Krakow, Poland
| | - Izabela Czyzynska-Cichon
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Karolina Matyjaszczyk-Gwarda
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
| | - Stefan Chlopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Bobrzynskiego 14, 30-348 Krakow, Poland; (B.P.); (A.B.); (A.K.-R.); (J.S.-P.); (E.B.); (K.C.); (Z.M.); (I.C.-C.); (G.K.); (K.M.-G.)
- Faculty of Pharmacology, Jagiellonian University Medical College, Grzegorzecka 16, 31-531 Krakow, Poland
- Correspondence:
| |
Collapse
|
15
|
Bubb KJ, Ravindran D, Cartland SP, Finemore M, Clayton ZE, Tsang M, Tang O, Kavurma MM, Patel S, Figtree GA. β 3 Adrenergic Receptor Stimulation Promotes Reperfusion in Ischemic Limbs in a Murine Diabetic Model. Front Pharmacol 2021; 12:666334. [PMID: 33967810 PMCID: PMC8100512 DOI: 10.3389/fphar.2021.666334] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Aims/Hypothesis: Peripheral arterial disease (PAD) is a major burden, resulting in limb claudication, repeated surgical interventions and amputation. There is an unmet need for improved medical management of PAD that improves quality of life, maintains activities of daily life and reduces complications. Nitric oxide (NO)/redox balance is a key regulator of angiogenesis. We have previously shown beneficial effects of a β3 adrenergic receptor (β3AR) agonist on NO/redox balance. We hypothesized that β3AR stimulation would have therapeutic potential in PAD by promoting limb angiogenesis. Methods: The effect of the β3AR agonist CL 316,243 (1–1,000 nmol/L in vitro, 1 mg/kg/day s. c) was tested in established angiogenesis assays with human endothelial cells and patient-derived endothelial colony forming cells. Post-ischemia reperfusion was determined in streptozotocin and/or high fat diet-induced diabetic and non-diabetic mice in vivo using the hind limb ischemia model. Results: CL 316,243 caused accelerated recovery from hind limb ischemia in non-diabetic and type 1 and 2 diabetic mice. Increased eNOS activity and decreased superoxide generation were detected in hind limb ischemia calf muscle from CL 316, 243 treated mice vs. controls. The protective effect of CL 316,243 in diabetic mice was associated with >50% decreases in eNOS glutathionylation and nitrotyrosine levels. The β3AR agonist directly promoted angiogenesis in endothelial cells in vitro. These pro-angiogenic effects were β3AR and NOS-dependent. Conclusion/Interpretation:β3AR stimulation increased angiogenesis in diabetic ischemic limbs, with demonstrable improvements in NO/redox balance and angiogenesis elicited by a selective agonist. The orally available β3AR agonist, Mirabegron, used for overactive bladder syndrome, makes translation to a clinical trial by repurposing of a β3AR agonist to target PAD immediately feasible.
Collapse
Affiliation(s)
- Kristen J Bubb
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia.,Department of Physiology, Biomedicine Discovery Institute, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, VIC, Australia
| | - Dhanya Ravindran
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Siân P Cartland
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Meghan Finemore
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Zoe E Clayton
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Michael Tsang
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Owen Tang
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| | - Mary M Kavurma
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Sanjay Patel
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Heart Research Institute, Eliza St Newtown, Sydney, NSW, Australia
| | - Gemma A Figtree
- University of Sydney, Faculty of Medicine and Health, Sydney, NSW, Australia.,Kolling Institute of Medical Research, Royal North Shore Hospital, St Leonards, NSW, Australia
| |
Collapse
|
16
|
Saunders SL, Hutchinson DS, Britton FC, Liu L, Markus I, Sandow SL, Murphy TV. Effect of β 1 /β 2 -adrenoceptor blockade on β 3 -adrenoceptor activity in the rat cremaster muscle artery. Br J Pharmacol 2021; 178:1789-1804. [PMID: 33506492 DOI: 10.1111/bph.15398] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/19/2021] [Accepted: 01/19/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE The physiological role of vascular β3 -adrenoceptors is not fully understood. Recent evidence suggests cardiac β3 -adrenoceptors are functionally effective after down-regulation of β1 /β2 -adrenoceptors. The functional interaction between the β3 -adrenoceptor and other β-adrenoceptor subtypes in rat striated muscle arteries was investigated. EXPERIMENTAL APPROACH Studies were performed in cremaster muscle arteries isolated from male Sprague-Dawley rats. β-adrenoceptor expression was assessed through RT-PCR and immunofluorescence. Functional effects of β3 -adrenoceptor agonists and antagonists and other β-adrenoceptor ligands were measured using pressure myography. KEY RESULTS All three β-adrenoceptor subtypes were present in the endothelium of the cremaster muscle artery. The β3 -adrenoceptor agonists mirabegron and CL 316,243 had no effect on the diameter of pressurized (70 mmHg) cremaster muscle arterioles with myogenic tone, while the β3 -adrenoceptor agonist SR 58611A and the nonselective β-adrenoceptor agonist isoprenaline caused concentration-dependent dilation. In the presence of β1/2 -adrenoceptor antagonists nadolol (10 μM), atenolol (1 μM) and ICI 118,551 (0.1 μM) both mirabegron and CL 316,243 were effective in causing vasodilation and the potency of SR 58611A was enhanced, while responses to isoprenaline were inhibited. The β3 -adrenoceptor antagonist L 748,337 (1 μM) inhibited vasodilation caused by β3 -adrenoceptor agonists (in the presence of β1/2 -adrenoceptor blockade), but L 748,337 had no effect on isoprenaline-induced vasodilation. CONCLUSION AND IMPLICATIONS All three β-adrenoceptor subtypes were present in the endothelium of the rat cremaster muscle artery, but β3 -adrenoceptor mediated vasodilation was only evident after blockade of β1/2 -adrenoceptors. This suggests constitutive β1/2 -adrenoceptor activity inhibits β3 -adrenoceptor function in the endothelium of skeletal muscle resistance arteries.
Collapse
Affiliation(s)
- Samantha L Saunders
- Physiology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Dana S Hutchinson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Fiona C Britton
- Department of Biomedical Sciences, School of Dental Medicine, University of Nevada, Las Vegas, Las Vegas, Nevada, USA
| | - Lu Liu
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Irit Markus
- Pharmacology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| | - Shaun L Sandow
- Physiology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia.,Biomedical Science, School of Health and Sports Science, University of the Sunshine Coast, Maroochydore, Queensland, Australia
| | - Timothy V Murphy
- Physiology, School of Medical Sciences, University of New South Wales, Sydney, New South Wales, Australia
| |
Collapse
|
17
|
Michel LYM, Farah C, Balligand JL. The Beta3 Adrenergic Receptor in Healthy and Pathological Cardiovascular Tissues. Cells 2020; 9:cells9122584. [PMID: 33276630 PMCID: PMC7761574 DOI: 10.3390/cells9122584] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 11/27/2020] [Accepted: 11/30/2020] [Indexed: 12/15/2022] Open
Abstract
The third isotype of beta-adrenoreceptors (β3-AR) has recently come (back) into focus after the observation of its expression in white and beige human adipocytes and its implication in metabolic regulation. This coincides with the recent development and marketing of agonists at the human receptor with superior specificity. Twenty years ago, however, we and others described the expression of β3-AR in human myocardium and its regulation of contractility and cardiac remodeling. Subsequent work from many laboratories has since expanded the characterization of β3-AR involvement in many aspects of cardiovascular physio(patho)logy, justifying the present effort to update current paradigms under the light of the most recent evidence.
Collapse
Affiliation(s)
- Lauriane Y. M. Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université Catholique de Louvain, B1.57.04, 57 Avenue Hippocrate, 1200 Brussels, Belgium; (L.Y.M.M.); (C.F.)
| | - Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université Catholique de Louvain, B1.57.04, 57 Avenue Hippocrate, 1200 Brussels, Belgium; (L.Y.M.M.); (C.F.)
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC), Université Catholique de Louvain, B1.57.04, 57 Avenue Hippocrate, 1200 Brussels, Belgium; (L.Y.M.M.); (C.F.)
- Department of Medicine, Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, 10 Avenue Hippocrate, 1200 Brussels, Belgium
- Correspondence: ; Tel.: +32-27645262
| |
Collapse
|
18
|
Wang Z, Li S, Wang R, Guo L, Xu D, Zhang T, Xu Y, Wang W, Wang M, Gan Z, Wang X. The protective effects of the β3 adrenergic receptor agonist BRL37344 against liver steatosis and inflammation in a rat model of high-fat diet-induced nonalcoholic fatty liver disease (NAFLD). Mol Med 2020; 26:54. [PMID: 32503411 PMCID: PMC7275314 DOI: 10.1186/s10020-020-00164-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Accepted: 03/30/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Our objective was to investigate the efficacy of the beta-3 adrenergic receptor (β3-AR) agonist BRL37344 for the prevention of liver steatosis and inflammation associated with nonalcoholic fatty liver disease (NAFLD). METHODS Four groups were established: a control group (given a standard diet), a high-fat diet (HFD) group, an HFD + β3-AR agonist (β3-AGO) group, and an HFD + β3-AR antagonist (β3-ANT) group. All rats were fed for 12 weeks. The β3-AR agonist BRL37344 and the antagonist L748337 were administered for the last 4 weeks with Alzet micro-osmotic pumps. The rat body weights (g) were measured at the end of the 4th, 8th, and 12th weeks. At the end of the 12th week, the liver weights were measured. Serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) were analyzed with a Hitachi automatic analyzer. The lipid levels of the triglycerides (TGs), total cholesterol (TC), and low-density lipoprotein cholesterol (LDL-C) and the concentrations of free fatty acids (FFAs) were also measured. An oil red O kit was used to detect lipid droplet accumulation in hepatocytes. Steatosis, ballooning degeneration and inflammation were histopathologically determined. The protein and mRNA expression levels of β3-AR, peroxisome proliferator-activated receptor-alpha (PPAR-α), peroxisome proliferator-activated receptor-gamma (PPAR-γ), mitochondrial carnitine palmitoyltransferase-1 (mCPT-1), and fatty acid translocase (FAT)/CD36 were measured by western blot analysis and RT-qPCR, respectively. RESULTS After treatment with the β3-AR agonist BRL37344 for 4 weeks, the levels of ALT, AST, TGs, TC, LDL-C and FFAs were decreased in the NAFLD model group compared with the HFD group. Body and liver weights, liver index values and lipid droplet accumulation were lower in the HFD + β3-AGO group than in the HFD group. Decreased NAFLD activity scores (NASs) also showed that liver steatosis and inflammation were ameliorated after treatment with BRL37344. Moreover, the β3-AR antagonist L748337 reversed these effects. Additionally, the protein and gene expression levels of β3-AR, PPAR-α, and mCPT-1 were increased in the HFD + β3-AGO group, whereas those of PPAR-γ and FAT/CD36 were decreased. CONCLUSION The β3-AR agonist BRL37344 is beneficial for reducing liver fat accumulation and for ameliorating liver steatosis and inflammation in NAFLD. These effects may be associated with PPARs/mCPT-1 and FAT/CD36.
Collapse
Affiliation(s)
- Ziwen Wang
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Shanshan Li
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Ruifeng Wang
- Gastroenterology Department, the Fourth Affiliated Hospital of Harbin Medical University, #37 Yiyuan Street, Harbin, 150001 Heilongjiang China
| | - Liansheng Guo
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Dan Xu
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| | - Tieyuan Zhang
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Yifan Xu
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Wenlong Wang
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Min Wang
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Zhongwei Gan
- Harbin Medical University, #157 Baojian Street, Harbin, 150081 Heilongjiang China
| | - Xiaobing Wang
- Gastroenterology Department, the First Affiliated Hospital of Harbin Medical University, #23 Postal Street, Harbin, 150001 Heilongjiang China
| |
Collapse
|
19
|
Abstract
Hypertension is still the number one global killer. No matter what causes are, lowering blood pressure can significantly reduce cardiovascular complications, cardiovascular death, and total death. Unfortunately, some hypertensive individuals simply do not know having hypertension. Some knew it but either not being treated or treated but blood pressure does not achieve goal. The reasons for inadequate control of blood pressure are many. One important reason is that we are not very familiar with antihypertensive agents and less attention has been paid to comorbidities, complications as well as the hypertension-modified target organ damage in patients with hypertension. The right antihypertensive drug was not given to the right hypertensive patients at right time. This reviewer studied comprehensively the literature, hopefully that the review will help improve antihypertensive drug selection and antihypertensive therapy.
Collapse
Affiliation(s)
- Rutai Hui
- Chinese Academy of Medical Sciences FUWAI Hospital Hypertension Division, 167 Beilishilu West City District, 100037, Beijing People's Republic of China, China.
| |
Collapse
|
20
|
Arioglu-Inan E, Kayki-Mutlu G, Michel MC. Cardiac β 3 -adrenoceptors-A role in human pathophysiology? Br J Pharmacol 2019; 176:2482-2495. [PMID: 30801686 DOI: 10.1111/bph.14635] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 01/11/2019] [Accepted: 01/29/2019] [Indexed: 01/06/2023] Open
Abstract
As β3 -adrenoceptors were first demonstrated to be expressed in adipose tissue they have received much attention for their metabolic effects in obesity and diabetes. After the existence of this subtype had been suggested to be present in the heart, studies focused on its role in cardiac function. While the presence and functional role of β3 -adrenoceptors in the heart has not uniformly been detected, there is a broad consensus that they become up-regulated in pathological conditions associated with increased sympathetic activity such as heart failure and diabetes. When detected, the β3 -adrenceptor has been demonstrated to mediate negative inotropic effects in an inhibitory G protein-dependent manner through the NO-cGMP-PKG signalling pathway. Whether these negative inotropic effects provide protection from the adverse effects induced by overstimulation of β1 /β2 -adrenoceptors or in themselves are potentially harmful is controversial, but ongoing clinical studies in patients with congestive heart failure are testing the hypothesis that β3 -adrenceptor agonism has a beneficial effect. LINKED ARTICLES: This article is part of a themed section on Adrenoceptors-New Roles for Old Players. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.14/issuetoc.
Collapse
Affiliation(s)
- Ebru Arioglu-Inan
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Gizem Kayki-Mutlu
- Department of Pharmacology, Faculty of Pharmacy, Ankara University, Ankara, Turkey
| | - Martin C Michel
- Department of Pharmacology, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
21
|
Everything You Always Wanted to Know about β 3-AR * (* But Were Afraid to Ask). Cells 2019; 8:cells8040357. [PMID: 30995798 PMCID: PMC6523418 DOI: 10.3390/cells8040357] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 03/26/2019] [Accepted: 04/12/2019] [Indexed: 12/22/2022] Open
Abstract
The beta-3 adrenergic receptor (β3-AR) is by far the least studied isotype of the beta-adrenergic sub-family. Despite its study being long hampered by the lack of suitable animal and cellular models and inter-species differences, a substantial body of literature on the subject has built up in the last three decades and the physiology of β3-AR is unraveling quickly. As will become evident in this work, β3-AR is emerging as an appealing target for novel pharmacological approaches in several clinical areas involving metabolic, cardiovascular, urinary, and ocular disease. In this review, we will discuss the most recent advances regarding β3-AR signaling and function and summarize how these findings translate, or may do so, into current clinical practice highlighting β3-AR’s great potential as a novel therapeutic target in a wide range of human conditions.
Collapse
|
22
|
Bubb KJ, Ritchie RH, Figtree GA. Modified redox signaling in vasculature after chronic infusion of the insulin receptor antagonist, S961. Microcirculation 2018; 26:e12501. [PMID: 30178465 DOI: 10.1111/micc.12501] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 07/02/2018] [Accepted: 08/30/2018] [Indexed: 11/28/2022]
Abstract
BACKGROUND Type 2 diabetes and associated vascular complications cause substantial morbidity and mortality. It is important to investigate mechanisms and test therapies in relevant physiological models, yet few animal models adequately recapitulate all aspects of the human condition. OBJECTIVE We sought to determine the potential of using an insulin receptor antagonist, S961, in mice for investigating vascular pathophysiology. METHODS S961 was infused into mice for 4 weeks. Blood glucose was monitored, and insulin was measured at the end of the protocol. Blood pressure and pressor responses to vasodilators were measured in cannulated mice, and vascular reactive oxygen and nitrogen species were measured in isolated tissue. RESULTS S961 infusion-induced hyperglycemia and hyperinsulinemia. There was evidence of increased vascular reactive oxygen and nitrogen species and modification of NO-mediated signaling. Pressor responses to a NO donor were attenuated, but responses to bradykinin were preserved. CONCLUSIONS Infusion of S961, an insulin receptor antagonist, results in the production of a mouse model of type 2 diabetes that may be useful for investigating redox signaling in the vasculature of insulin-resistant mice over the short term. It is limited by both the transient nature of the hyperglycemia and incomplete functional analogy to the human condition.
Collapse
Affiliation(s)
- Kristen J Bubb
- Cardiovascular and Thoracic Health, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Rebecca H Ritchie
- Heart Failure Pharmacology Laboratory, Basic Science Domain, Baker Heart and Diabetes Institute, Melbourne, Victoria, Australia
| | - Gemma A Figtree
- Cardiovascular and Thoracic Health, Kolling Institute of Medical Research, Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
23
|
Mirabegron, a β 3-adrenoceptor agonist reduced platelet aggregation through cyclic adenosine monophosphate accumulation. Eur J Pharmacol 2018; 829:79-84. [PMID: 29654782 DOI: 10.1016/j.ejphar.2018.04.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 11/22/2022]
Abstract
Mirabegron is a β3-adrenoceptor agonist and released on the marked for the treatment of overactive bladder. Because mirabegron is the only β3-adrenoceptor agonist available and substances that increase the levels of cyclic adenosine monophosphate (cAMP) inhibit platelet activity, we tested the hypothesis that mirabegron could have antiplatelet activity. Collagen- and thrombin induced platelet aggregation, thromboxane B2 (TXB2) and cyclic nucleotides quantification and calcium (Ca2+) mobilization were determined in the absence and presence of mirabegron in human washed platelets. Our results revealed that mirabegron (10-300 µM) produced significant inhibitions on platelet aggregation induced by collagen- or thrombin, accompanied by greater intracellular levels of cAMP. The β3-adrenoceptor antagonist L 748,337 (1 µM) and the adenylate cyclase inhibitor, SQ 22,536 (100 µM) reversed the inhibition induced by mirabegron in thrombin-stimulated platelets. The selective antagonists for β1-and β2-adrenoceptors, atenolol and ICI 117,551 (3 µM), respectively did not interfere on the inhibition induced by mirabegron. In Fluo-4 loaded platelets, mirabegron reduced the total and intracellular Ca2+ levels. Pre-incubation with mirabegron almost abolished the levels of TXB2. Mirabegron did not augment the intracellular levels of cyclic guanosine monophosphate. In conclusion, mirabegron inhibited human platelet aggregation through cAMP accumulation, thus suggesting that substances that activate β3-adrenoceptor could be beneficial as adjuvant antiplatelet therapy.
Collapse
|
24
|
Abstract
Nitric oxide (NO) signalling has pleiotropic roles in biology and a crucial function in cardiovascular homeostasis. Tremendous knowledge has been accumulated on the mechanisms of the nitric oxide synthase (NOS)-NO pathway, but how this highly reactive, free radical gas signals to specific targets for precise regulation of cardiovascular function remains the focus of much intense research. In this Review, we summarize the updated paradigms on NOS regulation, NO interaction with reactive oxidant species in specific subcellular compartments, and downstream effects of NO in target cardiovascular tissues, while emphasizing the latest developments of molecular tools and biomarkers to modulate and monitor NO production and bioavailability.
Collapse
Affiliation(s)
- Charlotte Farah
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| | - Jean-Luc Balligand
- Pole of Pharmacology and Therapeutics (FATH), Institut de Recherche Experimentale et Clinique (IREC) and Cliniques Universitaires Saint-Luc, Université Catholique de Louvain, UCL-FATH Tour Vésale 5th Floor, 52 Avenue Mounier B1.53.09, 1200 Brussels, Belgium
| |
Collapse
|
25
|
Boon EAJ, Croft KD, Shinde S, Hodgson JM, Ward NC. The acute effect of coffee on endothelial function and glucose metabolism following a glucose load in healthy human volunteers. Food Funct 2018; 8:3366-3373. [PMID: 28858362 DOI: 10.1039/c7fo00926g] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A diet rich in plant polyphenols has been suggested to reduce the incidence of cardiovascular disease and type 2 diabetes mellitus, in part, via improvements in endothelial function. Coffee is a rich source of phenolic compounds including the phenolic acid, chlorogenic acid (CGA). The aim of the study was to investigate the effect of coffee as a whole beverage on endothelial function, blood pressure and blood glucose concentration. Twelve healthy men and women were recruited to a randomised, placebo-controlled, cross-over study, with three treatments tested: (i) 18 g of ground caffeinated coffee containing 300 mg CGA in 200 mL of hot water, (ii) 18 g of decaffeinated coffee containing 287 mg CGA in 200 mL of hot water, and (iii) 200 mL of hot water (control). Treatment beverages were consumed twice, two hours apart, with the second beverage consumed simultaneously with a 75 g glucose load. Blood pressure was recorded and the finger prick glucose test was performed at time = 0 and then every 30 minutes up to 2 hours. Endothelial function, assessed using flow-mediated dilatation (FMD) of the brachial artery, was measured at 1 hour and a blood sample taken at 2 hours to measure plasma nitrate/nitrite and 5-CGA concentrations. The FMD response was significantly higher in the caffeinated coffee group compared to both decaffeinated coffee and water groups (P < 0.001). There was no significant difference in the FMD response between decaffeinated coffee and water. Blood glucose concentrations and blood pressure were not different between the three treatment groups. In conclusion, the consumption of caffeinated coffee resulted in a significant improvement in endothelial function, but there was no evidence for benefit regarding glucose metabolism or blood pressure. Although the mechanism has yet to be elucidated the results suggest that coffee as a whole beverage may improve endothelial function, or that caffeine is the component of coffee responsible for improving FMD.
Collapse
Affiliation(s)
- Evan A J Boon
- Schools of Medicine and Biomedical Science, University of Western Australia, Perth, Western Australia.
| | | | | | | | | |
Collapse
|
26
|
Hadi N, Al-Amran F, Al-Turfy M, Shaker S, Yousif N, Al-Aubaidy H. Response of internal mammary artery wall to L-carnitine in patients undergoing coronary artery bypass. VASCULAR INVESTIGATION AND THERAPY 2018. [DOI: 10.4103/vit.vit_11_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
27
|
Schiavone S, Camerino GM, Mhillaj E, Zotti M, Colaianna M, De Giorgi A, Trotta A, Cantatore FP, Conte E, Bove M, Tucci P, Morgese MG, Trabace L. Visceral Fat Dysfunctions in the Rat Social Isolation Model of Psychosis. Front Pharmacol 2017; 8:787. [PMID: 29167640 PMCID: PMC5682313 DOI: 10.3389/fphar.2017.00787] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2017] [Accepted: 10/17/2017] [Indexed: 01/18/2023] Open
Abstract
Medication with neuroleptics has been associated with adipose tissue dysfunctions and, in particular, with increased visceral fat amount. However, several studies suggested that antipsychotic treatment might not be the main responsible of fat mass accumulation, as this has been also described in not treated psychotic patients. One of the most used “drug-free” rodent models of psychosis is the social isolation rearing of young adult rats, which provides a non-pharmacologic method of inducing long-term alterations reminiscent of symptoms seen in psychotic patients. Recent data highlighted a crucial role of redox imbalance in adipose tissue dysfunctions, in terms of decreased antioxidant defense and increased reactive oxygen species (ROS). Here, we investigated possible oxidative stress-related biomolecular alterations associated with visceral fat increase in 7 week isolated rats. To this purpose, we quantified total and visceral fat amount by using dual-energy X-ray (DEXA) absorptiometry. On visceral fat, we analyzed the expression of specific ROS-producer genes (Nox1, Nox4, Hmox-1), antioxidant enzymes (Prdx1 and Ucp-1) and oxidative stress-induced damage markers (Cidea, Slc2a4, and Acacb). The impact of oxidative stress on beta3-adrenergic receptors (Adrb3), at both mRNA and protein level, was also assessed. We found that 7 weeks of social isolation induced an increase in total and visceral fat, associated with a decrease in Prdx1 (mRNA and protein) as well as Ucp-1 mRNA levels and an enhanced expression of Nox1 (mRNA and protein) and Hmox-1 mRNA. No differences were detected in Nox4 mRNA levels between grouped and isolated animals. Elevations in Cidea, Slc2a4, and Acacb expression in visceral fat of isolated animals accounted for oxidative stress-related damage in this tissue, further associated with a significant increase in Adrb3 mRNA and protein. Our results provide a novel understanding of the pathological link existing among psychosocial stress-induced psychosis, adipose tissue dysfunctions and redox imbalance, opening new therapeutic perspectives for the treatment of alterations in peripheral tissues associated with this mental disorder.
Collapse
Affiliation(s)
- Stefania Schiavone
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Giulia M Camerino
- Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Emanuela Mhillaj
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Margherita Zotti
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Marilena Colaianna
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Angelo De Giorgi
- Dual Diagnosis Unit, Azienda Sanitaria Locale della Provincia di Foggia, Foggia, Italy
| | - Antonello Trotta
- Rheumatology Unit, Foggia City Hospital "Ospedali Riuniti", Foggia, Italy
| | | | - Elena Conte
- Department of Pharmacy and Drug Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Maria Bove
- Department of Physiology and Pharmacology, Sapienza Università di Roma, Rome, Italy
| | - Paolo Tucci
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Maria G Morgese
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| | - Luigia Trabace
- Department of Clinical and Experimental Medicine, University of Foggia, Foggia, Italy
| |
Collapse
|
28
|
Song X, Yang B, Qiu F, Jia M, Fu G. High glucose and free fatty acids induce endothelial progenitor cell senescence via PGC-1α/SIRT1 signaling pathway. Cell Biol Int 2017; 41:1146-1159. [PMID: 28786152 DOI: 10.1002/cbin.10833] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Accepted: 07/28/2017] [Indexed: 01/01/2023]
Affiliation(s)
- Xiaoxiao Song
- The Department of Endocrinology; Second Affiliated Hospital, College of Medicine, Zhejiang University; Hangzhou 310009 Zhejiang Province China
- The Department of Cardiology; Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University; Hangzhou 310016 Zhejiang Province China
| | - Boyun Yang
- The Department of Endocrinology; Second Affiliated Hospital, College of Medicine, Zhejiang University; Hangzhou 310009 Zhejiang Province China
| | - Fuyu Qiu
- The Department of Cardiology; Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University; Hangzhou 310016 Zhejiang Province China
| | - Minyue Jia
- The Department of Endocrinology; Second Affiliated Hospital, College of Medicine, Zhejiang University; Hangzhou 310009 Zhejiang Province China
| | - Guosheng Fu
- The Department of Cardiology; Biomedical Research (Therapy) Center, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University; Hangzhou 310016 Zhejiang Province China
| |
Collapse
|
29
|
Bubb KJ, Birgisdottir AB, Tang O, Hansen T, Figtree GA. Redox modification of caveolar proteins in the cardiovascular system- role in cellular signalling and disease. Free Radic Biol Med 2017; 109:61-74. [PMID: 28188926 DOI: 10.1016/j.freeradbiomed.2017.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/18/2017] [Accepted: 02/05/2017] [Indexed: 02/07/2023]
Abstract
Rapid and coordinated release of a variety of reactive oxygen species (ROS) such as superoxide (O2.-), hydrogen peroxide (H2O2) and peroxynitrite, in specific microdomains, play a crucial role in cell signalling in the cardiovascular system. These reactions are mediated by reversible and functional modifications of a wide variety of key proteins. Dysregulation of this oxidative signalling occurs in almost all forms of cardiovascular disease (CVD), including at the very early phases. Despite the heavily publicized failure of "antioxidants" to improve CVD progression, pharmacotherapies such as those targeting the renin-angiotensin system, or statins, exert at least part of their large clinical benefit via modulating cellular redox signalling. Over 250 proteins, including receptors, ion channels and pumps, and signalling proteins are found in the caveolae. An increasing proportion of these are being recognized as redox regulated-proteins, that reside in the immediate vicinity of the two major cellular sources of ROS, nicotinamide adenine dinucleotide phosphate oxidase (Nox) and uncoupled endothelial nitric oxide synthase (eNOS). This review focuses on what is known about redox signalling within the caveolae, as well as endogenous protective mechanisms utilized by the cell, and new approaches to targeting dysregulated redox signalling in the caveolae as a therapeutic strategy in CVD.
Collapse
Affiliation(s)
- Kristen J Bubb
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Asa Birna Birgisdottir
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; Department of Cardiothoracic and Vascular Surgery, Heart and Lung Clinic, University Hospital of North Norway, Tromsø, Norway
| | - Owen Tang
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Thomas Hansen
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| |
Collapse
|
30
|
Bubb KJ, Kok C, Tang O, Rasko NB, Birgisdottir AB, Hansen T, Ritchie R, Bhindi R, Reisman SA, Meyer C, Ward K, Karimi Galougahi K, Figtree GA. The NRF2 activator DH404 attenuates adverse ventricular remodeling post-myocardial infarction by modifying redox signalling. Free Radic Biol Med 2017; 108:585-594. [PMID: 28438659 DOI: 10.1016/j.freeradbiomed.2017.04.027] [Citation(s) in RCA: 32] [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: 07/21/2016] [Revised: 03/24/2017] [Accepted: 04/19/2017] [Indexed: 12/29/2022]
Abstract
BACKGROUND The novel synthetic triterpenoid, bardoxolone methyl, has the ability to upregulate cytoprotective proteins via induction of the nuclear factor erythroid-2-related factor 2 (Nrf2) pathway. This makes it a promising therapeutic agent in disease states characterized by dysregulated oxidative signalling. We have examined the effect of a Nrf2 activator, dihydro-CDDO-trifluoroethyl amide (DH404), a derivative of bardoxolone methyl, on post-infarct cardiac remodeling in rats. METHODS/RESULTS DH404, administered from day 2 post myocardial infarction (MI: 30min transient ischemia followed by reperfusion) resulted in almost complete protection against adverse ventricular remodeling as assessed at day 28 (left ventricular end-systolic area: sham 0.14±0.01cm2, MI vehicle 0.29±0.04cm2 vs. MI DH404 0.18±0.02cm2, P<0.05); infarct size (21.3±3.4% MI vehicle vs. 10.9±2.3% MI DH404, P<0.05) with associated benefits on systolic function (fractional shortening: sham 71.9±2.6%, MI vehicle 36.2±1.9% vs. MI DH404 58.6±4.0%, P<0.05). These structural and functional benefits were associated with lower myocardial expression of atrial natriuretic peptide (ANP, P<0.01 vs. MI vehicle), and decreased fibronectin (P<0.01 vs. MI vehicle) in DH404-treated MI rats at 28 days. MI increased glutathionylation of endothelial nitric oxide synthase (eNOS) in vitro - a molecular switch that uncouples the enzyme, increasing superoxide production and decreasing nitric oxide (NO) bioavailability. MI-induced eNOS glutathionylation was substantially ameliorated by DH404. An associated increase in glutaredoxin 1 (Grx1) co-immunoprecipitation with eNOS without a change in expression was mechanistically intriguing. Indeed, in parallel in vitro experiments, silencing of Grx1 abolished the protective effect of DH404 against Angiotensin II-induced eNOS uncoupling. CONCLUSION The bardoxolone derivative DH404 significantly attenuated cardiac remodeling post MI, at least in part, by re-coupling of eNOS and increasing the functional interaction of Grx1 with eNOS. This agent may have clinical benefits protecting against post MI cardiomyopathy.
Collapse
Affiliation(s)
- Kristen J Bubb
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia
| | - Cindy Kok
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia
| | - Owen Tang
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia
| | - Nathalie B Rasko
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia
| | - Asa B Birgisdottir
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia; Department of Cardiothoracic and Vascular Surgery, Heart and Lung Clinic, University Hospital of North Norway, Tromsø, Norway
| | - Thomas Hansen
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia
| | - Rebecca Ritchie
- Baker IDI Heart and Diabetes Institute, Melbourne, Australia
| | - Ravinay Bhindi
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia; Department of Cardiology, Royal North Shore Hospital and University of Sydney, Australia
| | | | | | - Keith Ward
- Reata Pharmaceuticals, Inc. Irving, TX, USA
| | - Keyvan Karimi Galougahi
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia
| | - Gemma A Figtree
- North Shore Heart Research Group, Kolling Institute, University of Sydney and Royal North Shore Hospital, Sydney, Australia; Department of Cardiology, Royal North Shore Hospital and University of Sydney, Australia.
| |
Collapse
|
31
|
Altara R, Giordano M, Nordén ES, Cataliotti A, Kurdi M, Bajestani SN, Booz GW. Targeting Obesity and Diabetes to Treat Heart Failure with Preserved Ejection Fraction. Front Endocrinol (Lausanne) 2017; 8:160. [PMID: 28769873 PMCID: PMC5512012 DOI: 10.3389/fendo.2017.00160] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 06/23/2017] [Indexed: 12/12/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a major unmet medical need that is characterized by the presence of multiple cardiovascular and non-cardiovascular comorbidities. Foremost among these comorbidities are obesity and diabetes, which are not only risk factors for the development of HFpEF, but worsen symptoms and outcome. Coronary microvascular inflammation with endothelial dysfunction is a common denominator among HFpEF, obesity, and diabetes that likely explains at least in part the etiology of HFpEF and its synergistic relationship with obesity and diabetes. Thus, pharmacological strategies to supplement nitric oxide and subsequent cyclic guanosine monophosphate (cGMP)-protein kinase G (PKG) signaling may have therapeutic promise. Other potential approaches include exercise and lifestyle modifications, as well as targeting endothelial cell mineralocorticoid receptors, non-coding RNAs, sodium glucose transporter 2 inhibitors, and enhancers of natriuretic peptide protective NO-independent cGMP-initiated and alternative signaling, such as LCZ696 and phosphodiesterase-9 inhibitors. Additionally, understanding the role of adipokines in HFpEF may lead to new treatments. Identifying novel drug targets based on the shared underlying microvascular disease process may improve the quality of life and lifespan of those afflicted with both HFpEF and obesity or diabetes, or even prevent its occurrence.
Collapse
Affiliation(s)
- Raffaele Altara
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, Oslo, Norway
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- *Correspondence: Raffaele Altara,
| | - Mauro Giordano
- Department of Medical, Surgical, Neurological, Metabolic and Geriatrics Sciences, University of Campania “L. Vanvitelli”, Caserta, Italy
| | - Einar S. Nordén
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, Oslo, Norway
- Bjørknes College, Oslo, Norway
| | - Alessandro Cataliotti
- Institute for Experimental Medical Research, Oslo University Hospital and University of Oslo, Oslo, Norway
- KG Jebsen Center for Cardiac Research, Oslo, Norway
| | - Mazen Kurdi
- Faculty of Sciences, Department of Chemistry and Biochemistry, Lebanese University, Hadath, Lebanon
| | - Saeed N. Bajestani
- Department of Pathology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
- Department of Ophthalmology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| | - George W. Booz
- Department of Pharmacology and Toxicology, School of Medicine, University of Mississippi Medical Center, Jackson, MS, United States
| |
Collapse
|
32
|
Fu J, Han Y, Wang J, Liu Y, Zheng S, Zhou L, Jose PA, Zeng C. Irisin Lowers Blood Pressure by Improvement of Endothelial Dysfunction via AMPK-Akt-eNOS-NO Pathway in the Spontaneously Hypertensive Rat. J Am Heart Assoc 2016; 5:e003433. [PMID: 27912206 PMCID: PMC5210324 DOI: 10.1161/jaha.116.003433] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 09/06/2016] [Indexed: 12/13/2022]
Abstract
BACKGROUND Exercise is a major nonpharmacological treatment for hypertension, but its underlying mechanisms are still not completely elucidated. Irisin, a polypeptide containing 112 amino acids, which is secreted mainly by skeletal muscle cells during exercise, exerts a protective role in metabolic diseases, such as diabetes mellitus and obesity. Because of the close relationship between irisin and metabolic diseases, we hypothesized that irisin may play a role in the regulation of blood pressure. METHODS AND RESULTS Blood pressures of male Wistar-Kyoto (WKY) rats and spontaneously hypertensive rats (SHRs) were monitored through the carotid artery. Our study found that acute intravenous injection of irisin reduced blood pressure in SHRs, but not WKY rats. Irisin, by itself, had no direct vasorelaxing effect in phenylephrine-preconstricted mesenteric arteries from SHRs. However, irisin augmented the acetylcholine-induced vasorelaxation in mesenteric arteries from SHRs that could be reversed by Nω-nitro-l-arginine-methyl ester (L-NAME; 100 μmol/L), indicating a role of nitric oxide (NO) in this action. Indeed, irisin increased NO production and phosphorylation of endothelial nirtic oxide synthase (eNOS) in endothelial cells. 5'-AMP-activated protein kinase (AMPK) was involved in the vasorelaxing effect of irisin because compound C (20 μmol/L), an AMPK inhibitor, blocked the irisin-mediated increase in phosphorylation of eNOS and protein kinase B (Akt) in endothelial cells and vasodilation in mesenteric arteries. CONCLUSIONS We conclude that acute administration of irisin lowers blood pressure of SHRs by amelioration of endothelial dysfunction of the mesenteric artery through the AMPK-Akt-eNOS-NO signaling pathway.
Collapse
Affiliation(s)
- Jinjuan Fu
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Yu Han
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Jialiang Wang
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Yukai Liu
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Shuo Zheng
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Lin Zhou
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| | - Pedro A Jose
- Division of Renal Diseases & Hypertension, Department of Medicine, The George Washington University School of Medicine & Health Sciences, Washington, DC
| | - Chunyu Zeng
- Department of Cardiology, Daping Hospital, Chongqing Institute of Cardiology, The Third Military Medical University, Chongqing, China
| |
Collapse
|
33
|
Hainer V. Beta3-adrenoreceptor agonist mirabegron – a potential antiobesity drug? Expert Opin Pharmacother 2016; 17:2125-2127. [DOI: 10.1080/14656566.2016.1233177] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
34
|
Hansen T, Galougahi KK, Celermajer D, Rasko N, Tang O, Bubb KJ, Figtree G. Oxidative and nitrosative signalling in pulmonary arterial hypertension — Implications for development of novel therapies. Pharmacol Ther 2016; 165:50-62. [DOI: 10.1016/j.pharmthera.2016.05.005] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|