151 results on '"Montezano AC"'
Search Results
2. Interferon stimulated gene 15 pathway is a novel mediator of endothelial dysfunction and aneurysms development in angiotensin II infused mice through increased oxidative stress
- Author
-
González-Amor M, García-Redondo AB, Jorge I, Zalba G, Becares M, Ruiz-Rodríguez MJ, Rodríguez C, Bermeo H, Rodrigues-Díez R, Rios FJ, Montezano AC, Martínez-González J, Vázquez J, Redondo JM, Touyz RM, Guerra S, Salaices M, and Briones AM
- Abstract
AIMS: Interferon-stimulated gene 15 (ISG15) encodes an ubiquitin-like protein that induces a reversible post-translational modification (ISGylation) and can also be secreted as a free form. ISG15 plays an essential role as host-defense response to microbial infection; however, its contribution to vascular damage associated to hypertension is unknown. METHODS AND RESULTS: Bioinformatics identified ISG15 as a mediator of hypertension-associated vascular damage. ISG15 expression positively correlated with systolic and diastolic blood pressure and carotid intima-media thickness in human peripheral blood mononuclear cells. Consistently, Isg15 expression was enhanced in aorta from hypertension models and in angiotensinII (AngII)-treated vascular cells and macrophages. Proteomics revealed differential expression of proteins implicated in cardiovascular function, extracellular matrix and remodeling, and vascular redox state in aorta from AngII-infused ISG15-/- mice. Moreover, ISG15-/- mice were protected against AngII-induced hypertension, vascular stiffness, elastin remodeling, endothelial dysfunction, and expression of inflammatory and oxidative stress markers. Conversely, mice with excessive ISGylation (USP18C61A) show enhanced AngII-induced hypertension, vascular fibrosis, inflammation and reactive oxygen species (ROS) generation along with elastin breaks, aortic dilation and rupture. Accordingly, human and murine abdominal aortic aneurysms showed augmented ISG15 expression. Mechanistically, ISG15 induces vascular ROS production, while antioxidant treatment prevented ISG15-induced endothelial dysfunction and vascular remodeling. CONCLUSION: ISG15 is a novel mediator of vascular damage in hypertension through oxidative stress and inflammation. TRANSLATIONAL PERSPECTIVE: Recent evidence from randomized clinical trials demonstrate the effectiveness of specific anti-inflammatory treatments in cardiovascular prevention. In this study we have identified a new inflammatory mediator involved in vascular damage in experimental and human hypertension and aneurysms. We found that interferon stimulated gene 15 (ISG15) is increased at the vascular level in animal models of hypertension and aneurysms. More importantly, ISG15 correlates with human hypertension, vascular remodeling, and aneurysms presence. Underlying mechanisms responsible for vascular damage induced by ISG15 include oxidative and inflammation. Our results further support the role of inflammation in vascular damage in different cardiovascular pathologies.
- Published
- 2021
3. 35 Fetuin-A and toll-like receptor 4 regulate vascular function: role of Nox1
- Author
-
Neves, KB, primary, Lopes, RAM, additional, Leckerman, S, additional, Strembitska, A, additional, Jenkins, C, additional, Thomson, J, additional, Graham, D, additional, Touyz, RM, additional, and Montezano, AC, additional
- Published
- 2015
- Full Text
- View/download PDF
4. 39 Molecular mechanisms of VEGFR inhibition-induced endothelial cell damage
- Author
-
Rios, F, primary, Montezano, AC, additional, Van Der Mey, L, additional, Small, H, additional, Savoia, C, additional, and Touyz, RM, additional
- Published
- 2015
- Full Text
- View/download PDF
5. 32 Premature vascular ageing in hypertension: role of aldosterone and nadph oxidases
- Author
-
Harvey, AP, primary, Montezano, AC, additional, Hood, KY, additional, Lopes, RAM, additional, Ceravolo, GS, additional, Yabe-Nishimura, C, additional, Graham, D, additional, and Touyz, RM, additional
- Published
- 2015
- Full Text
- View/download PDF
6. 41 Vascular protein oxidation and redox proteomics in human hypertension
- Author
-
Tsiropoulou, S, primary, Montezano, AC, additional, Scott, A, additional, Burchmore, RJ, additional, and Touyz, RM, additional
- Published
- 2015
- Full Text
- View/download PDF
7. Increased endothelin-1 reactivity and endothelial dysfunction in carotid arteries from rats with hyperhomocysteinemia
- Author
-
De Andrade, CR, primary, Leite, PF, additional, Montezano, AC, additional, Casolari, DA, additional, Yogi, A, additional, Tostes, RC, additional, Haddad, R, additional, Eberlin, MN, additional, Laurindo, FRM, additional, De Souza, HP, additional, Corrêa, FMA, additional, and De Oliveira, AM, additional
- Published
- 2009
- Full Text
- View/download PDF
8. Testosterone induces vascular smooth muscle cell migration by NADPH oxidase and c-Src-dependent pathways.
- Author
-
Chignalia AZ, Schuldt EZ, Camargo LL, Montezano AC, Callera GE, Laurindo FR, Lopes LR, Avellar MC, Carvalho MH, Fortes ZB, Touyz RM, Tostes RC, Chignalia, Andreia Z, Schuldt, Elke Z, Camargo, Lívia L, Montezano, Augusto C, Callera, Gláucia E, Laurindo, Francisco R, Lopes, Lucia R, and Avellar, Maria Christina W
- Abstract
Testosterone has been implicated in vascular remodeling associated with hypertension. Molecular mechanisms underlying this are elusive, but oxidative stress may be important. We hypothesized that testosterone stimulates generation of reactive oxygen species (ROS) and migration of vascular smooth muscle cells (VSMCs), with enhanced effects in cells from spontaneously hypertensive rats (SHRs). The mechanisms (genomic and nongenomic) whereby testosterone induces ROS generation and the role of c-Src, a regulator of redox-sensitive migration, were determined. VSMCs from male Wistar-Kyoto rats and SHRs were stimulated with testosterone (10(-7) mol/L, 0-120 minutes). Testosterone increased ROS generation, assessed by dihydroethidium fluorescence and lucigenin-enhanced chemiluminescence (30 minutes [SHR] and 60 minutes [both strains]). Flutamide (androgen receptor antagonist) and actinomycin D (gene transcription inhibitor) diminished ROS production (60 minutes). Testosterone increased Nox1 and Nox4 mRNA levels and p47phox protein expression, determined by real-time PCR and immunoblotting, respectively. Flutamide, actinomycin D, and cycloheximide (protein synthesis inhibitor) diminished testosterone effects on p47phox. c-Src phosphorylation was observed at 30 minutes (SHR) and 120 minutes (Wistar-Kyoto rat). Testosterone-induced ROS generation was repressed by 3-(4-chlorophenyl) 1-(1,1-dimethylethyl)-1H-pyrazolo[3,4-day]pyrimidin-4-amine (c-Src inhibitor) in SHRs and reduced by apocynin (antioxidant/NADPH oxidase inhibitor) in both strains. Testosterone stimulated VSMCs migration, assessed by the wound healing technique, with greater effects in SHRs. Flutamide, apocynin, and 3-(4-chlorophenyl) 1-(1,1-dimethylethyl)-1H-pyrazolo[3,4-day]pyrimidin-4-amine blocked testosterone-induced VSMCs migration in both strains. Our study demonstrates that testosterone induces VSMCs migration via NADPH oxidase-derived ROS and c-Src-dependent pathways by genomic and nongenomic mechanisms, which are differentially regulated in VSMCs from Wistar-Kyoto rats and SHRs. [ABSTRACT FROM AUTHOR]
- Published
- 2012
- Full Text
- View/download PDF
9. Regulation of the novel Mg2+ transporter transient receptor potential melastatin 7 (TRPM7) cation channel by bradykinin in vascular smooth muscle cells.
- Author
-
Callera GE, He Y, Yogi A, Montezano AC, Paravicini T, Yao G, Touyz RM, Callera, Glaucia E, He, Ying, Yogi, Alvaro, Montezano, Augusto C, Paravicini, Tamara, Yao, Guoying, and Touyz, Rhian M
- Published
- 2009
- Full Text
- View/download PDF
10. Ang-(1-7) and ET-1 Interplay Through Mas and ET B Receptor Interaction Defines a Novel Vasoprotective Mechanism.
- Author
-
Montezano AC, Kuriakose J, Hood KY, Sin YY, Camargo LL, Namkung Y, Castro CH, Santos RA, Alves-Lopes R, Tejeda G, Passaglia P, Basheer S, Gallen E, Findlay JE, Awan FR, Laporte SA, MacLean MR, Baillie GS, and Touyz RM
- Abstract
Background: Ang-(1-7) (angiotensin (1-7)) via MasR (Mas receptor) opposes vaso-injurious actions of Ang II (angiotensin II) as shown in models of pulmonary hypertension. The underlying mechanisms remain unclear. We hypothesized cross talk between Ang-(1-7) and the protective arm of the ET-1 (endothelin-1) system involving MasR and ET
B R (endothelin receptor type B)., Methods/results: To address this, we studied multiple models: in vivo, in a mouse model of ET-1-associated vascular injury (hypoxia-induced pulmonary hypertension); ex vivo, in isolated mouse arteries; and in vitro, in human endothelial cells. Pulmonary hypertension mice exhibited pulmonary vascular remodeling, endothelial dysfunction, and ET-1-induced hypercontractility. Ang-(1-7) treatment (14 days) ameliorated these effects and increased the expression of vascular ETB R. In human endothelial cells, Ang-(1-7)-induced activation of eNOS (endothelial NO synthase)/NO was attenuated by A779 (MasR antagonist) and BQ788 (ETB R antagonist). A779 inhibited ET-1-induced signaling. Coimmunoprecipitation and peptide array experiments demonstrated the interaction between MasR and ETB R. Binding sites for ETB R were mapped to MasR (amino acids 290-314). Binding sites for MasR on ETB R were identified (amino acids 176-200). Peptides that disrupt MasR:ETB R prevented Ang-(1-7) and ET-1 signaling. Using high-throughput screening, we identified compounds that enhance MasR:ETB R interaction, which we termed enhancers. Enhancers increased Ang-(1-7)-induced eNOS activity, NO production, and Ang-(1-7)-mediated vasorelaxation, and reduced contractile responses., Conclusions: We identify cross talk between Ang-(1-7) and ET-1 through MasR:ETB R interaction as a novel network that is vasoprotective. Promoting coactivity between these systems amplifies Ang-(1-7) signaling, increases ET-1/ETB R-mediated vascular actions, and attenuates the injurious effects of ET-1. Enhancing Ang-(1-7)/MasR:ET-1/ETB R signaling may have therapeutic potential in conditions associated with vascular damage.- Published
- 2024
- Full Text
- View/download PDF
11. Pharmacological modulation of transglutaminase 2 in the unilateral ureteral obstruction mouse model.
- Author
-
Prat-Duran J, De Araujo IBBA, Juste N, Pinilla E, Rios FJ, Montezano AC, Touyz RM, Simonsen U, Nørregaard R, and Buus NH
- Subjects
- Animals, Mice, Male, Mice, Inbred C57BL, Blood Pressure drug effects, Tetrazoles pharmacology, Biphenyl Compounds pharmacology, Biphenyl Compounds therapeutic use, Collagen Type III metabolism, Collagen Type III genetics, Benzimidazoles pharmacology, Benzimidazoles therapeutic use, Fibronectins metabolism, Fibronectins genetics, Actins metabolism, Actins genetics, Transglutaminases metabolism, Transglutaminases genetics, Ureteral Obstruction drug therapy, Ureteral Obstruction pathology, Ureteral Obstruction complications, Protein Glutamine gamma Glutamyltransferase 2, GTP-Binding Proteins metabolism, GTP-Binding Proteins genetics, Disease Models, Animal, Fibrosis, Kidney drug effects, Kidney pathology, Kidney metabolism
- Abstract
Background: Transglutaminase 2 (TG2) is a multifunctional enzyme involved in fibrosis by promoting transforming-growth-factor-β1 and crosslinking of extracellular matrix proteins. These functions are dependent on the open conformation, while the closed state of TG2 can induce vasodilation. We explored the putative protective role of TG2 in its closed state on development of renal fibrosis and blood pressure (BP) regulation., Methods: We studied the unilateral ureteral obstruction (UUO) mouse model treated with LDN27219, which promotes the closed conformation of TG2. Mice were subjected to 7 days UUO or sham operation and treated with vehicle (n = 10), LDN27219 (15 mg/kg/12 h, n = 9) or candesartan (5 mg/kg/day, n = 10) as a clinically comparator. Renal expression of TG2 and pro-fibrotic mediators were evaluated by Western blotting, qPCR and histology, and BP by tail-cuff measurements., Results: Obstructed kidneys showed increased mRNA and protein expression of fibronectin, collagen 3α1 (Col3α1), α-smooth muscle actin and collagen staining. Despite increased renal TG2 mRNA, protein expression was reduced in all UUO groups, but with increased transamidase activity in the vehicle and candesartan groups. LDN27219 reduced mRNA expression of fibronectin and Col3α1, but their protein expression remained unchanged. In contrast to LDN27219, candesartan lowered BP without affecting expression of pro-fibrotic biomarkers., Conclusion: Renal TG2 mRNA and protein expression levels seem dissociated, with transamidase activity being increased. LDN27219 influences kidney pro-fibrotic markers at the mRNA level and attenuates transamidase activity but without affecting collagen content or BP. Our findings suggest that TG2 in its closed conformation has anti-fibrotic effects at the molecular level., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Estefano Pinilla reports financial support was provided by Erik Hørslev and Birgit Hørslev Fund. Niels Henrik Buus reports financial support was provided by Aarhus University Research Foundation. Aarhus University has patent #EP3607948A1 licensed to Estéfano Pinilla. Aarhus University has patent #EP3607948A1 licensed to Ulf Simonsen. If there are other authors, they declare that they have no known competing financial interests or personal relationships that could have appeared to influence the work reported in this paper., (Copyright © 2024 The Authors. Published by Elsevier B.V. All rights reserved.)
- Published
- 2024
- Full Text
- View/download PDF
12. Reactive oxygen species in hypertension.
- Author
-
Camargo LL, Rios FJ, Montezano AC, and Touyz RM
- Abstract
Hypertension is a leading risk factor for stroke, heart disease and chronic kidney disease. Multiple interacting factors and organ systems increase blood pressure and cause target-organ damage. Among the many molecular elements involved in the development of hypertension are reactive oxygen species (ROS), which influence cellular processes in systems that contribute to blood pressure elevation (such as the cardiovascular, renal, immune and central nervous systems, or the renin-angiotensin-aldosterone system). Dysregulated ROS production (oxidative stress) is a hallmark of hypertension in humans and experimental models. Of the many ROS-generating enzymes, NADPH oxidases are the most important in the development of hypertension. At the cellular level, ROS influence signalling pathways that define cell fate and function. Oxidative stress promotes aberrant redox signalling and cell injury, causing endothelial dysfunction, vascular damage, cardiovascular remodelling, inflammation and renal injury, which are all important in both the causes and consequences of hypertension. ROS scavengers reduce blood pressure in almost all experimental models of hypertension; however, clinical trials of antioxidants have yielded mixed results. In this Review, we highlight the latest advances in the understanding of the role and the clinical implications of ROS in hypertension. We focus on cellular sources of ROS, molecular mechanisms of oxidative stress and alterations in redox signalling in organ systems, and their contributions to hypertension., (© 2024. Springer Nature Limited.)
- Published
- 2024
- Full Text
- View/download PDF
13. Mechanisms of Vascular Inflammation and Potential Therapeutic Targets: A Position Paper From the ESH Working Group on Small Arteries.
- Author
-
Rios FJ, de Ciuceis C, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Lopreiato M, Mavraganis G, Mengozzi A, Montezano AC, Stavropoulos K, Winklewski PJ, Wolf J, Costantino S, Doumas M, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, and Virdis A
- Subjects
- Animals, Humans, Arteries metabolism, Cardiovascular Diseases metabolism, Epigenesis, Genetic, Oxidative Stress physiology, Signal Transduction physiology, Vasculitis metabolism, Vasculitis immunology, Inflammation metabolism, Inflammation immunology
- Abstract
Inflammatory responses in small vessels play an important role in the development of cardiovascular diseases, including hypertension, stroke, and small vessel disease. This involves various complex molecular processes including oxidative stress, inflammasome activation, immune-mediated responses, and protein misfolding, which together contribute to microvascular damage. In addition, epigenetic factors, including DNA methylation, histone modifications, and microRNAs influence vascular inflammation and injury. These phenomena may be acquired during the aging process or due to environmental factors. Activation of proinflammatory signaling pathways and molecular events induce low-grade and chronic inflammation with consequent cardiovascular damage. Identifying mechanism-specific targets might provide opportunities in the development of novel therapeutic approaches. Monoclonal antibodies targeting inflammatory cytokines and epigenetic drugs, show promise in reducing microvascular inflammation and associated cardiovascular diseases. In this article, we provide a comprehensive discussion of the complex mechanisms underlying microvascular inflammation and offer insights into innovative therapeutic strategies that may ameliorate vascular injury in cardiovascular disease., Competing Interests: Disclosures None.
- Published
- 2024
- Full Text
- View/download PDF
14. SEC31A may be associated with pituitary hormone deficiency and gonadal dysgenesis.
- Author
-
Tobias ES, Lucas-Herald AK, Sagar D, Montezano AC, Rios FJ, De Lucca Camargo L, Hamilton G, Gazdagh G, Diver LA, Williams N, Herzyk P, Touyz RM, Greenfield A, McGowan R, and Ahmed SF
- Subjects
- Animals, Child, Child, Preschool, Female, Humans, Male, Mice, Mice, Knockout, Pedigree, Pituitary Hormones deficiency, Pituitary Hormones genetics, Vesicular Transport Proteins genetics, Gonadal Dysgenesis genetics, Hypopituitarism genetics, Hypopituitarism metabolism
- Abstract
Purpose: Disorders/differences of sex development (DSD) result from variants in many different human genes but, frequently, have no detectable molecular cause., Methods: Detailed clinical and genetic phenotyping was conducted on a family with three children. A Sec31a animal model and functional studies were used to investigate the significance of the findings., Results: By trio whole-exome DNA sequencing we detected a heterozygous de novo nonsense SEC31A variant, in three children of healthy non-consanguineous parents. The children had different combinations of disorders that included complete gonadal dysgenesis and multiple pituitary hormone deficiency. SEC31A encodes a component of the COPII coat protein complex, necessary for intracellular anterograde vesicle-mediated transport between the endoplasmic reticulum (ER) and Golgi. CRISPR-Cas9 targeted knockout of the orthologous Sec31a gene region resulted in early embryonic lethality in homozygous mice. mRNA expression of ER-stress genes ATF4 and CHOP was increased in the children, suggesting defective protein transport. The pLI score of the gene, from gnomAD data, is 0.02., Conclusions: SEC31A might underlie a previously unrecognised clinical syndrome comprising gonadal dysgenesis, multiple pituitary hormone deficiencies, dysmorphic features and developmental delay. However, a variant that remains undetected, in a different gene, may alternatively be causal in this family., (© 2024. The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2024
- Full Text
- View/download PDF
15. Effects of Sex Hormones on Vascular Reactivity in Boys With Hypospadias.
- Author
-
Lucas-Herald AK, Montezano AC, Alves-Lopes R, Haddow L, O'Toole S, Flett M, Lee B, Amjad SB, Steven M, McNeilly J, Brooksbank K, Touyz RM, and Ahmed SF
- Subjects
- Male, Humans, Infant, Nitroprusside pharmacology, Testosterone pharmacology, Estradiol pharmacology, Androgens pharmacology, Dihydrotestosterone pharmacology, Acetylcholine, Hypospadias surgery
- Abstract
Background: Arteries from boys with hypospadias demonstrate hypercontractility and impaired vasorelaxation. The role of sex hormones in these responses in unclear., Aims: We compared effects of sex steroids on vascular reactivity in healthy boys and boys with hypospadias., Methods: Excess foreskin tissue was obtained from 11 boys undergoing hypospadias repair (cases) and 12 undergoing routine circumcision (controls) (median age [range], 1.5 [1.2-2.7] years) and small resistance arteries were isolated. Vessels were mounted on wire myographs and vascular reactivity was assessed in the absence/presence of 17β-estradiol, dihydrotestosterone (DHT), and testosterone., Results: In controls, testosterone and 17β-estradiol increased contraction (percent of maximum contraction [Emax]: 83.74 basal vs 125.4 after testosterone, P < .0002; and 83.74 vs 110.2 after estradiol, P = .02). 17β-estradiol reduced vasorelaxation in arteries from controls (Emax: 10.6 vs 15.6 to acetylcholine, P < .0001; and Emax: 14.6 vs 20.5 to sodium nitroprusside, P < .0001). In hypospadias, testosterone (Emax: 137.9 vs 107.2, P = .01) and 17β-estradiol (Emax: 156.9 vs 23.6, P < .0001) reduced contraction. Androgens, but not 17β-estradiol, increased endothelium-dependent and endothelium-independent vasorelaxation in cases (Emax: 77.3 vs 51.7 with testosterone, P = .02; and vs 48.2 with DHT to acetylcholine, P = .0001; Emax: 43.0 vs 39.5 with testosterone, P = .02; and 39.6 vs 37.5 with DHT to sodium nitroprusside, P = .04)., Conclusion: In healthy boys, testosterone and 17β-estradiol promote a vasoconstrictor phenotype, whereas in boys with hypospadias, these sex hormones reduce vasoconstriction, with androgens promoting vasorelaxation. Differences in baseline artery function may therefore be sex hormone-independent and the impact of early-life variations in androgen exposure on vascular function needs further study., (© The Author(s) 2023. Published by Oxford University Press on behalf of the Endocrine Society.)
- Published
- 2024
- Full Text
- View/download PDF
16. Oxidative Stress and Endoplasmic Reticular Stress Interplay in the Vasculopathy of Hypertension.
- Author
-
Camargo LL, Wang Y, Rios FJ, McBride M, Montezano AC, and Touyz RM
- Subjects
- Humans, Reactive Oxygen Species metabolism, Endoplasmic Reticulum Stress genetics, Oxidation-Reduction, Oxidative Stress, Hypertension
- Abstract
Under physiologic conditions, reactive oxygen species (ROS) function as signalling molecules that control cell function. However, in pathologic conditions, increased generation of ROS triggers oxidative stress, which plays a role in vascular changes associated with hypertension, including endothelial dysfunction, vascular reactivity, and arterial remodelling (termed the vasculopathy of hypertension). The major source of ROS in the vascular system is NADPH oxidase (NOX). Increased NOX activity drives vascular oxidative stress in hypertension. Molecular mechanisms underlying vascular damage in hypertension include activation of redox-sensitive signalling pathways, post-translational modification of proteins, and oxidative damage of DNA and cytoplasmic proteins. In addition, oxidative stress leads to accumulation of proteins in the endoplasmic reticulum (ER) (termed ER stress), with consequent activation of the unfolded protein response (UPR). ER stress is emerging as a potential player in hypertension as abnormal protein folding in the ER leads to oxidative stress and dysregulated activation of the UPR promotes inflammation and injury in vascular and cardiac cells. In addition, the ER engages in crosstalk with exogenous sources of ROS, such as mitochondria and NOX, which can amplify redox processes. Here we provide an update of the role of ROS and NOX in hypertension and discuss novel concepts on the interplay between oxidative stress and ER stress., (Copyright © 2023 Canadian Cardiovascular Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
17. Recent Advances in Understanding the Mechanistic Role of Transient Receptor Potential Ion Channels in Patients With Hypertension.
- Author
-
Rios FJ, Sarafian RD, Camargo LL, Montezano AC, and Touyz RM
- Subjects
- Humans, Endothelial Cells metabolism, Muscle, Smooth, Vascular metabolism, Ions metabolism, Transient Receptor Potential Channels metabolism, Hypertension
- Abstract
The transient receptor potential (TRP) channel superfamily is a group of nonselective cation channels that function as cellular sensors for a wide range of physical, chemical, and environmental stimuli. According to sequence homology, TRP channels are categorized into 6 subfamilies: TRP canonical, TRP vanilloid, TRP melastatin, TRP ankyrin, TRP mucolipin, and TRP polycystin. They are widely expressed in different cell types and tissues and have essential roles in various physiological and pathological processes by regulating the concentration of ions (Ca
2+ , Mg2+ , Na+ , and K+ ) and influencing intracellular signalling pathways. Human data and experimental models indicate the importance of TRP channels in vascular homeostasis and hypertension. Furthermore, TRP channels have emerged as key players in oxidative stress and inflammation, important in the pathophysiology of cardiovascular diseases, including hypertension. In this review, we present an overview of the TRP channels with a focus on their role in hypertension. In particular, we highlight mechanisms activated by TRP channels in vascular smooth muscle and endothelial cells and discuss their contribution to processes underlying vascular dysfunction in hypertension., (Copyright © 2023 Canadian Cardiovascular Society. Published by Elsevier Inc. All rights reserved.)- Published
- 2023
- Full Text
- View/download PDF
18. Exosomes and the cardiovascular system: role in cardiovascular health and disease.
- Author
-
Neves KB, Rios FJ, Sevilla-Montero J, Montezano AC, and Touyz RM
- Subjects
- Humans, Proteins, Biomarkers metabolism, Lipids, Exosomes metabolism, MicroRNAs genetics, MicroRNAs metabolism, Cardiovascular System metabolism, Extracellular Vesicles metabolism
- Abstract
Exosomes, which are membrane-bound extracellular vesicles (EVs), are generated in the endosomal compartment of almost all eukaryotic cells. They are formed upon the fusion of multivesicular bodies and the plasma membrane and carry proteins, nucleic acids, lipids and other cellular constituents from their parent cells. Multiple factors influence their production including cell stress and injury, humoral factors, circulating toxins, and oxidative stress. They play an important role in intercellular communication, through their ability to transfer their cargo (proteins, lipids, RNAs) from one cell to another. Exosomes have been implicated in the pathophysiology of various diseases including cardiovascular disease (CVD), cancer, kidney disease, and inflammatory conditions. In addition, circulating exosomes may act as biomarkers for diagnostic and prognostic strategies for several pathological processes. In particular exosome-containing miRNAs have been suggested as biomarkers for the diagnosis and prognosis of myocardial injury, stroke and endothelial dysfunction. They may also have therapeutic potential, acting as vectors to deliver therapies in a targeted manner, such as the delivery of protective miRNAs. Transfection techniques are in development to load exosomes with desired cargo, such as proteins or miRNAs, to achieve up-regulation in the host cell or tissue. These advances in the field have the potential to assist in the detection and monitoring progress of a disease in patients during its early clinical stages, as well as targeted drug delivery., (© 2022 The Authors. The Journal of Physiology published by John Wiley & Sons Ltd on behalf of The Physiological Society.)
- Published
- 2023
- Full Text
- View/download PDF
19. The importance of microvascular inflammation in ageing and age-related diseases: a position paper from the ESH working group on small arteries, section of microvascular inflammation.
- Author
-
Mengozzi A, de Ciuceis C, Dell'oro R, Georgiopoulos G, Lazaridis A, Nosalski R, Pavlidis G, Tual-Chalot S, Agabiti-Rosei C, Anyfanti P, Camargo LL, Dąbrowska E, Quarti-Trevano F, Hellmann M, Masi S, Mavraganis G, Montezano AC, Rios FJ, Winklewski PJ, Wolf J, Costantino S, Gkaliagkousi E, Grassi G, Guzik TJ, Ikonomidis I, Narkiewicz K, Paneni F, Rizzoni D, Stamatelopoulos K, Stellos K, Taddei S, Touyz RM, Triantafyllou A, and Virdis A
- Subjects
- Humans, Chronic Disease, Microcirculation, Inflammation, Arteries
- Abstract
Microcirculation is pervasive and orchestrates a profound regulatory cross-talk with the surrounding tissue and organs. Similarly, it is one of the earliest biological systems targeted by environmental stressors and consequently involved in the development and progression of ageing and age-related disease. Microvascular dysfunction, if not targeted, leads to a steady derangement of the phenotype, which cumulates comorbidities and eventually results in a nonrescuable, very high-cardiovascular risk. Along the broad spectrum of pathologies, both shared and distinct molecular pathways and pathophysiological alteration are involved in the disruption of microvascular homeostasis, all pointing to microvascular inflammation as the putative primary culprit. This position paper explores the presence and the detrimental contribution of microvascular inflammation across the whole spectrum of chronic age-related diseases, which characterise the 21st-century healthcare landscape. The manuscript aims to strongly affirm the centrality of microvascular inflammation by recapitulating the current evidence and providing a clear synoptic view of the whole cardiometabolic derangement. Indeed, there is an urgent need for further mechanistic exploration to identify clear, very early or disease-specific molecular targets to provide an effective therapeutic strategy against the otherwise unstoppable rising prevalence of age-related diseases., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
20. Impact of Environmental Factors on Hypertension and Associated Cardiovascular Disease.
- Author
-
Rios FJ, Montezano AC, Camargo LL, and Touyz RM
- Subjects
- Humans, Life Style, Mediastinum, Risk Factors, Cardiovascular Diseases epidemiology, Cardiovascular Diseases etiology, Hypertension epidemiology, Hypertension etiology
- Abstract
Hypertension is the primary cause of cardiovascular diseases and is responsible for nearly 9 million deaths worldwide annually. Increasing evidence indicates that in addition to pathophysiologic processes, numerous environmental factors, such as geographic location, lifestyle choices, socioeconomic status, and cultural practices, influence the risk, progression, and severity of hypertension, even in the absence of genetic risk factors. In this review, we discuss the impact of some environmental determinants on hypertension. We focus on clinical data from large population studies and discuss some potential molecular and cellular mechanisms. We highlight how these environmental determinants are interconnected, as small changes in one factor might affect others, and further affect cardiovascular health. In addition, we discuss the crucial impact of socioeconomic factors and how these determinants influence diverse communities with economic disparities. Finally, we address opportunities and challenges for new research to address gaps in knowledge on understanding molecular mechanisms whereby environmental factors influence development of hypertension and associated cardiovascular disease., (Copyright © 2023 Canadian Cardiovascular Society. Published by Elsevier Inc. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
21. Vasoprotective effects of NOX4 are mediated via polymerase and transient receptor potential melastatin 2 cation channels in endothelial cells.
- Author
-
Alves-Lopes R, Lacchini S, Neves KB, Harvey A, Montezano AC, and Touyz RM
- Subjects
- Animals, Mice, Rats, Calcium metabolism, Endothelial Cells metabolism, Hydrogen Peroxide pharmacology, Nitric Oxide metabolism, Poly(ADP-ribose) Polymerase Inhibitors, Hypertension metabolism, TRPM Cation Channels genetics, TRPM Cation Channels metabolism
- Abstract
Background: NOX4 activation has been implicated to have vasoprotective and blood pressure (BP)-lowering effects. Molecular mechanisms underlying this are unclear, but NOX4-induced regulation of the redox-sensitive Ca 2+ channel TRPM2 and effects on endothelial nitric oxide synthase (eNOS)-nitric oxide signalling may be important., Method: Wild-type and LinA3, renin-expressing hypertensive mice, were crossed with NOX4 knockout mice. Vascular function was measured by myography. Generation of superoxide (O 2- ) and hydrogen peroxide (H 2 O 2 ) were assessed by lucigenin and amplex red, respectively, and Ca 2+ influx by Cal-520 fluorescence in rat aortic endothelial cells (RAEC)., Results: BP was increased in NOX4KO, LinA3 and LinA3/NOX4KO mice. This was associated with endothelial dysfunction and vascular remodelling, with exaggerated effects in NOX4KO groups. The TRPM2 activator, ADPR, improved vascular relaxation in LinA3/NOX4KO mice, an effect recapitulated by H 2 O 2 . Inhibition of PARP and TRPM2 with olaparib and 2-APB, respectively, recapitulated endothelial dysfunction in NOX4KO. In endothelial cells, Ang II increased H 2 O 2 generation and Ca 2+ influx, effects reduced by TRPM2 siRNA, TRPM2 inhibitors (8-br-cADPR, 2-APB), olaparib and GKT137831 (NOX4 inhibitor). Ang II-induced eNOS activation was blocked by NOX4 and TRPM2 siRNA, GKT137831, PEG-catalase and 8-br-cADPR., Conclusion: Our findings indicate that NOX4-induced H 2 O 2 production activates PARP/TRPM2, Ca 2+ influx, eNOS activation and nitric oxide release in endothelial cells. NOX4 deficiency impairs Ca 2+ homeostasis leading to endothelial dysfunction, an effect exacerbated in hypertension. We define a novel pathway linking endothelial NOX4/H 2 O 2 to eNOS/nitric oxide through PARP/TRPM2/Ca 2+ . This vasoprotective pathway is perturbed when NOX4 is downregulated and may have significance in conditions associated with endothelial dysfunction, including hypertension., (Copyright © 2023 The Author(s). Published by Wolters Kluwer Health, Inc.)
- Published
- 2023
- Full Text
- View/download PDF
22. SARS-CoV-2 spike protein induces endothelial inflammation via ACE2 independently of viral replication.
- Author
-
Montezano AC, Camargo LL, Mary S, Neves KB, Rios FJ, Stein R, Lopes RA, Beattie W, Thomson J, Herder V, Szemiel AM, McFarlane S, Palmarini M, and Touyz RM
- Subjects
- Humans, Spike Glycoprotein, Coronavirus, Angiotensin-Converting Enzyme 2, SARS-CoV-2, Inflammation, Virus Replication, RNA, Double-Stranded, Endothelial Cells, COVID-19
- Abstract
COVID-19, caused by SARS-CoV-2, is a respiratory disease associated with inflammation and endotheliitis. Mechanisms underling inflammatory processes are unclear, but angiotensin converting enzyme 2 (ACE2), the receptor which binds the spike protein of SARS-CoV-2 may be important. Here we investigated whether spike protein binding to ACE2 induces inflammation in endothelial cells and determined the role of ACE2 in this process. Human endothelial cells were exposed to SARS-CoV-2 spike protein, S1 subunit (rS1p) and pro-inflammatory signaling and inflammatory mediators assessed. ACE2 was modulated pharmacologically and by siRNA. Endothelial cells were also exposed to SARS-CoV-2. rSP1 increased production of IL-6, MCP-1, ICAM-1 and PAI-1, and induced NFkB activation via ACE2 in endothelial cells. rS1p increased microparticle formation, a functional marker of endothelial injury. ACE2 interacting proteins involved in inflammation and RNA biology were identified in rS1p-treated cells. Neither ACE2 expression nor ACE2 enzymatic function were affected by rSP1. Endothelial cells exposed to SARS-CoV-2 virus did not exhibit viral replication. We demonstrate that rSP1 induces endothelial inflammation via ACE2 through processes that are independent of ACE2 enzymatic activity and viral replication. We define a novel role for ACE2 in COVID-19- associated endotheliitis., (© 2023. Springer Nature Limited.)
- Published
- 2023
- Full Text
- View/download PDF
23. Notch3/Hes5 Induces Vascular Dysfunction in Hypoxia-Induced Pulmonary Hypertension Through ER Stress and Redox-Sensitive Pathways.
- Author
-
Morris HE, Neves KB, Nilsen M, Montezano AC, MacLean MR, and Touyz RM
- Subjects
- Animals, Mice, Basic Helix-Loop-Helix Transcription Factors metabolism, Hypoxia metabolism, Myocytes, Smooth Muscle metabolism, Oxidation-Reduction, Pulmonary Artery metabolism, Reactive Oxygen Species metabolism, Receptor, Notch3 genetics, Receptor, Notch3 metabolism, Repressor Proteins metabolism, Humans, Hypertension, Hypertension, Pulmonary metabolism, Pulmonary Arterial Hypertension metabolism
- Abstract
Background: Notch3 (neurogenic locus notch homolog protein 3) is implicated in vascular diseases, including pulmonary hypertension (PH)/pulmonary arterial hypertension. However, molecular mechanisms remain elusive. We hypothesized increased Notch3 activation induces oxidative and endoplasmic reticulum (ER) stress and downstream redox signaling, associated with procontractile pulmonary artery state, pulmonary vascular dysfunction, and PH development., Methods: Studies were performed in TgNotch3
R169C mice (harboring gain-of-function [GOF] Notch3 mutation) exposed to chronic hypoxia to induce PH, and examined by hemodynamics. Molecular and cellular studies were performed in pulmonary artery smooth muscle cells from pulmonary arterial hypertension patients and in mouse lung. Notch3-regulated genes/proteins, ER stress, ROCK (Rho-associated kinase) expression/activity, Ca2+ transients and generation of reactive oxygen species, and nitric oxide were measured. Pulmonary vascular reactivity was assessed in the presence of fasudil (ROCK inhibitor) and 4-phenylbutyric acid (ER stress inhibitor)., Results: Hypoxia induced a more severe PH phenotype in TgNotch3R169C mice versus controls. TgNotch3R169C mice exhibited enhanced Notch3 activation and expression of Notch3 targets Hes Family BHLH Transcription Factor 5 (Hes5), with increased vascular contraction and impaired vasorelaxation that improved with fasudil/4-phenylbutyric acid. Notch3 mutation was associated with increased pulmonary vessel Ca2+ transients, ROCK activation, ER stress, and increased reactive oxygen species generation, with reduced NO generation and blunted sGC (soluble guanylyl cyclase)/cGMP signaling. These effects were ameliorated by N-acetylcysteine. pulmonary artery smooth muscle cells from patients with pulmonary arterial hypertension recapitulated Notch3/Hes5 signaling, ER stress and redox changes observed in PH mice., Conclusions: Notch3 GOF amplifies vascular dysfunction in hypoxic PH. This involves oxidative and ER stress, and ROCK. We highlight a novel role for Notch3/Hes5-redox signaling and important interplay between ER and oxidative stress in PH., Competing Interests: Disclosures None.- Published
- 2023
- Full Text
- View/download PDF
24. Vascular mechanisms of post-COVID-19 conditions: Rho-kinase is a novel target for therapy.
- Author
-
Sykes RA, Neves KB, Alves-Lopes R, Caputo I, Fallon K, Jamieson NB, Kamdar A, Legrini A, Leslie H, McIntosh A, McConnachie A, Morrow A, McFarlane RW, Mangion K, McAbney J, Montezano AC, Touyz RM, Wood C, and Berry C
- Subjects
- Humans, Female, Middle Aged, Male, rho-Associated Kinases metabolism, 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid pharmacology, Post-Acute COVID-19 Syndrome, Cardiovascular Diseases diagnosis, Cardiovascular Diseases drug therapy, COVID-19
- Abstract
Background: In post-coronavirus disease-19 (post-COVID-19) conditions (long COVID), systemic vascular dysfunction is implicated, but the mechanisms are uncertain, and the treatment is imprecise., Methods and Results: Patients convalescing after hospitalization for COVID-19 and risk factor matched controls underwent multisystem phenotyping using blood biomarkers, cardiorenal and pulmonary imaging, and gluteal subcutaneous biopsy (NCT04403607). Small resistance arteries were isolated and examined using wire myography, histopathology, immunohistochemistry, and spatial transcriptomics. Endothelium-independent (sodium nitroprusside) and -dependent (acetylcholine) vasorelaxation and vasoconstriction to the thromboxane A2 receptor agonist, U46619, and endothelin-1 (ET-1) in the presence or absence of a RhoA/Rho-kinase inhibitor (fasudil), were investigated. Thirty-seven patients, including 27 (mean age 57 years, 48% women, 41% cardiovascular disease) 3 months post-COVID-19 and 10 controls (mean age 57 years, 20% women, 30% cardiovascular disease), were included. Compared with control responses, U46619-induced constriction was increased (P = 0.002) and endothelium-independent vasorelaxation was reduced in arteries from COVID-19 patients (P < 0.001). This difference was abolished by fasudil. Histopathology revealed greater collagen abundance in COVID-19 arteries {Masson's trichrome (MT) 69.7% [95% confidence interval (CI): 67.8-71.7]; picrosirius red 68.6% [95% CI: 64.4-72.8]} vs. controls [MT 64.9% (95% CI: 59.4-70.3) (P = 0.028); picrosirius red 60.1% (95% CI: 55.4-64.8), (P = 0.029)]. Greater phosphorylated myosin light chain antibody-positive staining in vascular smooth muscle cells was observed in COVID-19 arteries (40.1%; 95% CI: 30.9-49.3) vs. controls (10.0%; 95% CI: 4.4-15.6) (P < 0.001). In proof-of-concept studies, gene pathways associated with extracellular matrix alteration, proteoglycan synthesis, and viral mRNA replication appeared to be upregulated., Conclusion: Patients with post-COVID-19 conditions have enhanced vascular fibrosis and myosin light change phosphorylation. Rho-kinase activation represents a novel therapeutic target for clinical trials., (© The Author(s) 2023. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2023
- Full Text
- View/download PDF
25. Role of PARP and TRPM2 in VEGF Inhibitor-Induced Vascular Dysfunction.
- Author
-
Neves KB, Alves-Lopes R, Montezano AC, and Touyz RM
- Subjects
- Humans, Mice, Animals, Reactive Oxygen Species metabolism, Vascular Endothelial Growth Factor A metabolism, Poly(ADP-ribose) Polymerase Inhibitors pharmacology, Poly(ADP-ribose) Polymerase Inhibitors metabolism, Poly(ADP-ribose) Polymerase Inhibitors therapeutic use, Axitinib therapeutic use, Endothelial Cells metabolism, Nitric Oxide metabolism, Angiogenesis Inhibitors, TRPM Cation Channels genetics, Antineoplastic Agents therapeutic use, Neoplasms drug therapy, Hypertension drug therapy
- Abstract
Background Hypertension and vascular toxicity are major unwanted side effects of antiangiogenic drugs, such as vascular endothelial growth factor inhibitors (VEGFis), which are effective anticancer drugs but have unwanted side effects, including vascular toxicity and hypertension. Poly (ADP-ribose) polymerase (PARP) inhibitors, used to treat ovarian and other cancers, have also been associated with elevated blood pressure. However, when patients with cancer receive both olaparib, a PARP inhibitor, and VEGFi, the risk of blood pressure elevation is reduced. Underlying molecular mechanisms are unclear, but PARP-regulated transient receptor potential cation channel, subfamily M, member 2 (TRPM2), a redox-sensitive calcium channel, may be important. We investigated whether PARP/TRPM2 plays a role in VEGFi-induced vascular dysfunction and whether PARP inhibition ameliorates the vasculopathy associated with VEGF inhibition. Methods and Results Human vascular smooth muscle cells (VSMCs), human aortic endothelial cells, and wild-type mouse mesenteric arteries were studied. Cells/arteries were exposed to axitinib (VEGFi) alone and in combination with olaparib. Reactive oxygen species production, Ca
2+ influx, protein/gene analysis, PARP activity, and TRPM2 signaling were assessed in VSMCs, and nitric oxide levels were determined in endothelial cells. Vascular function was assessed by myography. Axitinib increased PARP activity in VSMCs in a reactive oxygen species-dependent manner. Endothelial dysfunction and hypercontractile responses were ameliorated by olaparib and a TRPM2 blocker (8-Br-cADPR). VSMC reactive oxygen species production, Ca2+ influx, and phosphorylation of myosin light chain 20 and endothelial nitric oxide synthase (Thr495 ) were augmented by axitinib and attenuated by olaparib and TRPM2 inhibition. Proinflammatory markers were upregulated in axitinib-stimulated VSMCs, which was reduced by reactive oxygen species scavengers and PARP-TRPM2 inhibition. Human aortic endothelial cells exposed to combined olaparib and axitinib showed nitric oxide levels similar to VEGF-stimulated cells. Conclusions Axitinib-mediated vascular dysfunction involves PARP and TRPM2, which, when inhibited, ameliorate the injurious effects of VEGFi. Our findings define a potential mechanism whereby PARP inhibitor may attenuate vascular toxicity in VEGFi-treated patients with cancer.- Published
- 2023
- Full Text
- View/download PDF
26. Interferon-stimulated gene 15 pathway is a novel mediator of endothelial dysfunction and aneurysms development in angiotensin II infused mice through increased oxidative stress.
- Author
-
González-Amor M, García-Redondo AB, Jorge I, Zalba G, Becares M, Ruiz-Rodríguez MJ, Rodríguez C, Bermeo H, Rodrigues-Díez R, Rios FJ, Montezano AC, Martínez-González J, Vázquez J, Redondo JM, Touyz RM, Guerra S, Salaices M, and Briones AM
- Subjects
- Mice, Humans, Animals, Elastin metabolism, Reactive Oxygen Species metabolism, Angiotensin II metabolism, Interferons metabolism, Leukocytes, Mononuclear metabolism, Carotid Intima-Media Thickness, Oxidative Stress, Oxidation-Reduction, Inflammation, Mice, Inbred C57BL, Hypertension chemically induced, Hypertension genetics, Hypertension metabolism, Aortic Aneurysm, Abdominal chemically induced, Aortic Aneurysm, Abdominal genetics, Aortic Aneurysm, Abdominal prevention & control
- Abstract
Aims: Interferon-stimulated gene 15 (ISG15) encodes a ubiquitin-like protein that induces a reversible post-translational modification (ISGylation) and can also be secreted as a free form. ISG15 plays an essential role as host-defence response to microbial infection; however, its contribution to vascular damage associated with hypertension is unknown., Methods and Results: Bioinformatics identified ISG15 as a mediator of hypertension-associated vascular damage. ISG15 expression positively correlated with systolic and diastolic blood pressure and carotid intima-media thickness in human peripheral blood mononuclear cells. Consistently, Isg15 expression was enhanced in aorta from hypertension models and in angiotensin II (AngII)-treated vascular cells and macrophages. Proteomics revealed differential expression of proteins implicated in cardiovascular function, extracellular matrix and remodelling, and vascular redox state in aorta from AngII-infused ISG15-/- mice. Moreover, ISG15-/- mice were protected against AngII-induced hypertension, vascular stiffness, elastin remodelling, endothelial dysfunction, and expression of inflammatory and oxidative stress markers. Conversely, mice with excessive ISGylation (USP18C61A) show enhanced AngII-induced hypertension, vascular fibrosis, inflammation and reactive oxygen species (ROS) generation along with elastin breaks, aortic dilation, and rupture. Accordingly, human and murine abdominal aortic aneurysms showed augmented ISG15 expression. Mechanistically, ISG15 induces vascular ROS production, while antioxidant treatment prevented ISG15-induced endothelial dysfunction and vascular remodelling., Conclusion: ISG15 is a novel mediator of vascular damage in hypertension through oxidative stress and inflammation., Competing Interests: Conflict of interest: none declared., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2021. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2022
- Full Text
- View/download PDF
27. TRPM7 deficiency exacerbates cardiovascular and renal damage induced by aldosterone-salt.
- Author
-
Rios FJ, Zou ZG, Harvey AP, Harvey KY, Camargo LL, Neves KB, Nichol SEF, Alves-Lopes R, Cheah A, Zahraa M, Ryazanov AG, Ryazanova L, Gudermann T, Chubanov V, Montezano AC, and Touyz RM
- Subjects
- Aldosterone pharmacology, Animals, Fibrosis, Kidney metabolism, Magnesium metabolism, Mice, Protein Phosphatase 2C metabolism, Sodium Chloride, Hyperaldosteronism genetics, Hyperaldosteronism metabolism, TRPM Cation Channels deficiency, TRPM Cation Channels genetics, TRPM Cation Channels metabolism
- Abstract
Hyperaldosteronism causes cardiovascular disease as well as hypomagnesemia. Mechanisms are ill-defined but dysregulation of TRPM7, a Mg
2+ -permeable channel/α-kinase, may be important. We examined the role of TRPM7 in aldosterone-dependent cardiovascular and renal injury by studying aldosterone-salt treated TRPM7-deficient (TRPM7+/Δkinase ) mice. Plasma/tissue [Mg2+ ] and TRPM7 phosphorylation were reduced in vehicle-treated TRPM7+/Δkinase mice, effects recapitulated in aldosterone-salt-treated wild-type mice. Aldosterone-salt treatment exaggerated vascular dysfunction and amplified cardiovascular and renal fibrosis, with associated increased blood pressure in TRPM7+/Δkinase mice. Tissue expression of Mg2+ -regulated phosphatases (PPM1A, PTEN) was downregulated and phosphorylation of Smad3, ERK1/2, and Stat1 was upregulated in aldosterone-salt TRPM7-deficient mice. Aldosterone-induced phosphorylation of pro-fibrotic signaling was increased in TRPM7+/Δkinase fibroblasts, effects ameliorated by Mg2+ supplementation. TRPM7 deficiency amplifies aldosterone-salt-induced cardiovascular remodeling and damage. We identify TRPM7 downregulation and associated hypomagnesemia as putative molecular mechanisms underlying deleterious cardiovascular and renal effects of hyperaldosteronism., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
28. Differential effects of cyclo-oxygenase 1 and 2 inhibition on angiogenesis inhibitor-induced hypertension and kidney damage.
- Author
-
Mirabito Colafella KM, van Dorst DCH, Neuman RI, Doorn LV, Neves KB, Montezano AC, Garrelds IM, van Veghel R, de Vries R, Uijl E, Clahsen-van Groningen MC, Baelde HJ, van den Meiracker AH, Touyz RM, Visser W, Danser AHJ, and Versmissen J
- Subjects
- Angiogenesis Inhibitors therapeutic use, Animals, Aspirin pharmacology, Cyclooxygenase 1 metabolism, Cyclooxygenase 2 metabolism, Endothelin-1 metabolism, Epoprostenol metabolism, Epoprostenol pharmacology, Epoprostenol therapeutic use, Female, Humans, Kidney metabolism, Pregnancy, Prostaglandin-Endoperoxide Synthases metabolism, Rats, Vascular Endothelial Growth Factor A metabolism, Hypertension chemically induced, Hypertension drug therapy, Hypertension metabolism, Pre-Eclampsia chemically induced, Pre-Eclampsia drug therapy, Pre-Eclampsia metabolism
- Abstract
Vascular endothelial growth factor antagonism with angiogenesis inhibitors in cancer patients induces a 'preeclampsia-like' syndrome including hypertension, proteinuria and elevated endothelin (ET)-1. Cyclo-oxygenase (COX) inhibition with aspirin is known to prevent the onset of preeclampsia in high-risk patients. In the present study, we hypothesised that treatment with aspirin would prevent the development of angiogenesis inhibitor-induced hypertension and kidney damage. Our aims were to compare the effects of low-dose (COX-1 inhibition) and high-dose (dual COX-1 and COX-2 inhibition) aspirin on blood pressure, vascular function, oxidative stress, ET-1 and prostanoid levels and kidney damage during angiogenesis-inhibitor therapy in rodents. To this end, Wistar Kyoto rats were treated with vehicle, angiogenesis inhibitor (sunitinib) alone or in combination with low- or high-dose aspirin for 8 days (n=5-7/group). Our results demonstrated that prostacyclin (PGI2) and ET-1 were increased during angiogenesis-inhibitor therapy, while thromboxane (TXA2) was unchanged. Both low- and high-dose aspirin blunted angiogenesis inhibitor-induced hypertension and vascular superoxide production to a similar extent, whereas only high-dose aspirin prevented albuminuria. While circulating TXA2 and prostaglandin F2α levels were reduced by both low- and high-dose aspirin, circulating and urinary levels PGI2 were only reduced by high-dose aspirin. Lastly, treatment with aspirin did not significantly affect ET-1 or vascular function. Collectively our findings suggest that prostanoids contribute to the development of angiogenesis inhibitor-induced hypertension and renal damage and that targeting the prostanoid pathway could be an effective strategy to mitigate the unwanted cardiovascular and renal toxicities associated with angiogenesis inhibitors., (© 2022 The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
29. Central role of c-Src in NOX5- mediated redox signalling in vascular smooth muscle cells in human hypertension.
- Author
-
Camargo LL, Montezano AC, Hussain M, Wang Y, Zou Z, Rios FJ, Neves KB, Alves-Lopes R, Awan FR, Guzik TJ, Jensen T, Hartley RC, and Touyz RM
- Subjects
- Actins metabolism, Angiotensin II metabolism, Animals, Cells, Cultured, Humans, Melitten metabolism, Melitten pharmacology, Mice, Mice, Transgenic, Myocytes, Smooth Muscle metabolism, NADPH Oxidase 5 genetics, NADPH Oxidase 5 metabolism, NADPH Oxidase 5 pharmacology, Oxidation-Reduction, Protein-Tyrosine Kinases metabolism, RNA, Small Interfering genetics, Reactive Oxygen Species metabolism, Hypertension, Muscle, Smooth, Vascular metabolism
- Abstract
Aims: NOX-derived reactive oxygen species (ROS) are mediators of signalling pathways implicated in vascular smooth muscle cell (VSMC) dysfunction in hypertension. Among the numerous redox-sensitive kinases important in VSMC regulation is c-Src. However, mechanisms linking NOX/ROS to c-Src are unclear, especially in the context of oxidative stress in hypertension. Here, we investigated the role of NOX-induced oxidative stress in VSMCs in human hypertension focusing on NOX5, and explored c-Src, as a putative intermediate connecting NOX5-ROS to downstream effector targets underlying VSMC dysfunction., Methods and Results: VSMC from arteries from normotensive (NT) and hypertensive (HT) subjects were studied. NOX1,2,4,5 expression, ROS generation, oxidation/phosphorylation of signalling molecules, and actin polymerization and migration were assessed in the absence and presence of NOX5 (melittin) and Src (PP2) inhibitors. NOX5 and p22phox-dependent NOXs (NOX1-4) were down-regulated using NOX5 siRNA and p22phox-siRNA approaches. As proof of concept in intact vessels, vascular function was assessed by myography in transgenic mice expressing human NOX5 in a VSMC-specific manner. In HT VSMCs, NOX5 was up-regulated, with associated oxidative stress, hyperoxidation (c-Src, peroxiredoxin, DJ-1), and hyperphosphorylation (c-Src, PKC, ERK1/2, MLC20) of signalling molecules. NOX5 siRNA reduced ROS generation in NT and HT subjects. NOX5 siRNA, but not p22phox-siRNA, blunted c-Src phosphorylation in HT VSMCs. NOX5 siRNA reduced phosphorylation of MLC20 and FAK in NT and HT. In p22phox- silenced HT VSMCs, Ang II-induced phosphorylation of MLC20 was increased, effects blocked by melittin and PP2. NOX5 and c-Src inhibition attenuated actin polymerization and migration in HT VSMCs. In NOX5 transgenic mice, vascular hypercontractilty was decreased by melittin and PP2., Conclusion: We define NOX5/ROS/c-Src as a novel feedforward signalling network in human VSMCs. Amplification of this system in hypertension contributes to VSMC dysfunction. Dampening the NOX5/ROS/c-Src pathway may ameliorate hypertension-associated vascular injury., (© The Author(s) 2021. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2022
- Full Text
- View/download PDF
30. Sex steroids receptors, hypertension, and vascular ageing.
- Author
-
Connelly PJ, Casey H, Montezano AC, Touyz RM, and Delles C
- Subjects
- Adolescent, Aging, Aldosterone metabolism, Female, Humans, Male, Renin-Angiotensin System physiology, Hypertension, Receptors, Steroid metabolism
- Abstract
Sex hormone receptors are expressed throughout the vasculature and play an important role in the modulation of blood pressure in health and disease. The functions of these receptors may be important in the understanding of sexual dimorphism observed in the pathophysiology of both hypertension and vascular ageing. The interconnectivity of these factors can be exemplified in postmenopausal females, who with age and estrogen deprivation, surpass males with regard to hypertension prevalence, despite experiencing significantly less disease burden in their estrogen replete youth. Estrogen and androgen receptors mediate their actions via direct genomic effects or rapid non-genomic signaling, involving a host of mediators. The expression and subtype composition of these receptors changes through the lifespan in response to age, disease and hormonal exposure. These factors may promote sex steroid receptor-mediated alterations to the Renin-Angiotensin-Aldosterone System (RAAS), and increases in oxidative stress and inflammation, thereby contributing to the development of hypertension and vascular injury with age., (© 2021. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
31. Peptides derived from the SARS-CoV-2 receptor binding motif bind to ACE2 but do not block ACE2-mediated host cell entry or pro-inflammatory cytokine induction.
- Author
-
Mahindra A, Tejeda G, Rossi M, Janha O, Herbert I, Morris C, Morgan DC, Beattie W, Montezano AC, Hudson B, Tobin AB, Bhella D, Touyz RM, Jamieson AG, Baillie GS, and Blair CM
- Subjects
- A549 Cells, Humans, Protein Interaction Domains and Motifs, Angiotensin-Converting Enzyme 2 immunology, Antiviral Agents pharmacology, Peptides pharmacology, Protein Binding drug effects, Spike Glycoprotein, Coronavirus immunology, Virus Internalization
- Abstract
SARS-CoV-2 viral attachment and entry into host cells is mediated by a direct interaction between viral spike glycoproteins and membrane bound angiotensin-converting enzyme 2 (ACE2). The receptor binding motif (RBM), located within the S1 subunit of the spike protein, incorporates the majority of known ACE2 contact residues responsible for high affinity binding and associated virulence. Observation of existing crystal structures of the SARS-CoV-2 receptor binding domain (SRBD)-ACE2 interface, combined with peptide array screening, allowed us to define a series of linear native RBM-derived peptides that were selected as potential antiviral decoy sequences with the aim of directly binding ACE2 and attenuating viral cell entry. RBM1 (16mer): S443KVGGNYNYLYRLFRK458, RBM2A (25mer): E484GFNCYFPLQSYGFQPTNGVGYQPY508, RBM2B (20mer): F456NCYFPLQSYGFQPTNGVGY505 and RBM2A-Sc (25mer): NYGLQGSPFGYQETPYPFCNFVQYG. Data from fluorescence polarisation experiments suggested direct binding between RBM peptides and ACE2, with binding affinities ranging from the high nM to low μM range (Kd = 0.207-1.206 μM). However, the RBM peptides demonstrated only modest effects in preventing SRBD internalisation and showed no antiviral activity in a spike protein trimer neutralisation assay. The RBM peptides also failed to suppress S1-protein mediated inflammation in an endogenously expressing ACE2 human cell line. We conclude that linear native RBM-derived peptides are unable to outcompete viral spike protein for binding to ACE2 and therefore represent a suboptimal approach to inhibiting SARS-CoV-2 viral cell entry. These findings reinforce the notion that larger biologics (such as soluble ACE2, 'miniproteins', nanobodies and antibodies) are likely better suited as SARS-CoV-2 cell-entry inhibitors than short-sequence linear peptides., Competing Interests: The authors declare no competing interests.
- Published
- 2021
- Full Text
- View/download PDF
32. Osteoprotegerin regulates vascular function through syndecan-1 and NADPH oxidase-derived reactive oxygen species.
- Author
-
Alves-Lopes R, Neves KB, Strembitska A, Harvey AP, Harvey KY, Yusuf H, Haniford S, Hepburn RT, Dyet J, Beattie W, Haddow L, McAbney J, Graham D, and Montezano AC
- Subjects
- Animals, Cells, Cultured, Male, Mesenteric Arteries drug effects, Mesenteric Arteries enzymology, Mesenteric Arteries physiopathology, Mice, Inbred C57BL, Muscle, Smooth, Vascular enzymology, Muscle, Smooth, Vascular physiopathology, Myocytes, Smooth Muscle enzymology, NADPH Oxidase 1 genetics, NADPH Oxidase 1 metabolism, NADPH Oxidase 4 genetics, NADPH Oxidase 4 metabolism, NADPH Oxidases genetics, Rats, Inbred WKY, Signal Transduction, Mice, Rats, Hemodynamics drug effects, Muscle, Smooth, Vascular drug effects, Myocytes, Smooth Muscle drug effects, NADPH Oxidases metabolism, Osteoprotegerin toxicity, Oxidative Stress, Reactive Oxygen Species metabolism, Syndecan-1 metabolism
- Abstract
Osteogenic factors, such as osteoprotegerin (OPG), are protective against vascular calcification. However, OPG is also positively associated with cardiovascular damage, particularly in pulmonary hypertension, possibly through processes beyond effects on calcification. In the present study, we focused on calcification-independent vascular effects of OPG through activation of syndecan-1 and NADPH oxidases (Noxs) 1 and 4. Isolated resistance arteries from Wistar-Kyoto (WKY) rats, exposed to exogenous OPG, studied by myography exhibited endothelial and smooth muscle dysfunction. OPG decreased nitric oxide (NO) production, eNOS activation and increased reactive oxygen species (ROS) production in endothelial cells. In VSMCs, OPG increased ROS production, H2O2/peroxynitrite levels and activation of Rho kinase and myosin light chain. OPG vascular and redox effects were also inhibited by the syndecan-1 inhibitor synstatin (SSNT). Additionally, heparinase and chondroitinase abolished OPG effects on VSMCs-ROS production, confirming syndecan-1 as OPG molecular partner and suggesting that OPG binds to heparan/chondroitin sulphate chains of syndecan-1. OPG-induced ROS production was abrogated by NoxA1ds (Nox1 inhibitor) and GKT137831 (dual Nox1/Nox4 inhibitor). Tempol (SOD mimetic) inhibited vascular dysfunction induced by OPG. In addition, we studied arteries from Nox1 and Nox4 knockout (KO) mice. Nox1 and Nox4 KO abrogated OPG-induced vascular dysfunction. Vascular dysfunction elicited by OPG is mediated by a complex signalling cascade involving syndecan-1, Nox1 and Nox4. Our data identify novel molecular mechanisms beyond calcification for OPG, which may underlie vascular injurious effects of osteogenic factors in conditions such as hypertension and/or diabetes., (© 2021 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
33. Cardiovascular and Renal Risk Factors and Complications Associated With COVID-19.
- Author
-
Touyz RM, Boyd MOE, Guzik T, Padmanabhan S, McCallum L, Delles C, Mark PB, Petrie JR, Rios F, Montezano AC, Sykes R, and Berry C
- Abstract
The current COVID-19 pandemic, caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) virus, represents the largest medical challenge in decades. It has exposed unexpected cardiovascular vulnerabilities at all stages of the disease (pre-infection, acute phase, and subsequent chronic phase). The major cardiometabolic drivers identified as having epidemiologic and mechanistic associations with COVID-19 are abnormal adiposity, dysglycemia, dyslipidemia, and hypertension. Hypertension is of particular interest, because components of the renin-angiotensin system (RAS), which are critically involved in the pathophysiology of hypertension, are also implicated in COVID-19. Specifically, angiotensin-converting enzyme-2 (ACE2), a multifunctional protein of the RAS, which is part of the protective axis of the RAS, is also the receptor through which SARS-CoV-2 enters host cells, causing viral infection. Cardiovascular and cardiometabolic comorbidities not only predispose people to COVID-19, but also are complications of SARS-CoV-2 infection. In addition, increasing evidence indicates that acute kidney injury is common in COVID-19, occurs early and in temporal association with respiratory failure, and is associated with poor prognosis, especially in the presence of cardiovascular risk factors. Here, we discuss cardiovascular and kidney disease in the context of COVID-19 and provide recent advances on putative pathophysiological mechanisms linking cardiovascular disease and COVID-19, focusing on the RAS and ACE2, as well as the immune system and inflammation. We provide up-to-date information on the relationships among hypertension, diabetes, and COVID-19 and emphasize the major cardiovascular diseases associated with COVID-19. We also briefly discuss emerging cardiovascular complications associated with long COVID-19, notably postural tachycardia syndrome (POTS)., (© 2021 The Authors.)
- Published
- 2021
- Full Text
- View/download PDF
34. Selective Inhibition of the C-Domain of ACE (Angiotensin-Converting Enzyme) Combined With Inhibition of NEP (Neprilysin): A Potential New Therapy for Hypertension.
- Author
-
Alves-Lopes R, Montezano AC, Neves KB, Harvey A, Rios FJ, Skiba DS, Arendse LB, Guzik TJ, Graham D, Poglitsch M, Sturrock E, and Touyz RM
- Subjects
- Aminobutyrates therapeutic use, Angiotensin-Converting Enzyme Inhibitors pharmacology, Animals, Antihypertensive Agents therapeutic use, Biphenyl Compounds therapeutic use, Body Weight drug effects, Hypertension metabolism, Lisinopril therapeutic use, Liver drug effects, Liver metabolism, Mice, Mice, Transgenic, Pyridines pharmacology, Pyridines therapeutic use, Renin metabolism, Thiazepines pharmacology, Thiazepines therapeutic use, Aminobutyrates pharmacology, Antihypertensive Agents pharmacology, Biphenyl Compounds pharmacology, Blood Pressure drug effects, Hypertension drug therapy, Lisinopril pharmacology, Neprilysin antagonists & inhibitors
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
35. Lysophosphatidylcholine induces oxidative stress in human endothelial cells via NOX5 activation - implications in atherosclerosis.
- Author
-
da Silva JF, Alves JV, Silva-Neto JA, Costa RM, Neves KB, Alves-Lopes R, Carmargo LL, Rios FJ, Montezano AC, Touyz RM, and Tostes RC
- Subjects
- Atherosclerosis pathology, Calcium metabolism, Calcium Signaling, Cell Adhesion, Cells, Cultured, Coculture Techniques, Endothelial Cells enzymology, Endothelial Cells pathology, Enzyme Activation, Enzyme Inhibitors pharmacology, Humans, Intercellular Adhesion Molecule-1 genetics, Intercellular Adhesion Molecule-1 metabolism, Monocytes metabolism, NADPH Oxidase 5 antagonists & inhibitors, NADPH Oxidase 5 genetics, RNA Interference, Atherosclerosis enzymology, Endothelial Cells drug effects, Lysophosphatidylcholines toxicity, NADPH Oxidase 5 metabolism, Oxidative Stress drug effects, Reactive Oxygen Species metabolism
- Abstract
Objective: The mechanisms involved in NOX5 activation in atherosclerotic processes are not completely understood. The present study tested the hypothesis that lysophosphatidylcholine (LPC), a proatherogenic component of oxLDL, induces endothelial calcium influx, which drives NOX5-dependent reactive oxygen species (ROS) production, oxidative stress, and endothelial cell dysfunction., Approach: Human aortic endothelial cells (HAEC) were stimulated with LPC (10-5 M, for different time points). Pharmacological inhibition of NOX5 (Melittin, 10-7 M) and NOX5 gene silencing (siRNA) was used to determine the role of NOX5-dependent ROS production in endothelial oxidative stress induced by LPC. ROS production was determined by lucigenin assay and electron paramagnetic spectroscopy (EPR), calcium transients by Fluo4 fluorimetry, and NOX5 activity and protein expression by pharmacological assays and immunoblotting, respectively., Results: LPC increased ROS generation in endothelial cells at short (15 min) and long (4 h) stimulation times. LPC-induced ROS was abolished by a selective NOX5 inhibitor and by NOX5 siRNA. NOX1/4 dual inhibition and selective NOX1 inhibition only decreased ROS generation at 4 h. LPC increased HAEC intracellular calcium, important for NOX5 activation, and this was blocked by nifedipine and thapsigargin. Bapta-AM, selective Ca2+ chelator, prevented LPC-induced ROS production. NOX5 knockdown decreased LPC-induced ICAM-1 mRNA expression and monocyte adhesion to endothelial cells., Conclusion: These results suggest that NOX5, by mechanisms linked to increased intracellular calcium, is key to early LPC-induced endothelial oxidative stress and pro-inflammatory processes. Since these are essential events in the formation and progression of atherosclerotic lesions, the present study highlights an important role for NOX5 in atherosclerosis., (© 2021 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
36. Assessment and pathophysiology of microvascular disease: recent progress and clinical implications.
- Author
-
Masi S, Rizzoni D, Taddei S, Widmer RJ, Montezano AC, Lüscher TF, Schiffrin EL, Touyz RM, Paneni F, Lerman A, Lanza GA, and Virdis A
- Subjects
- Humans, Oxidative Stress, Cardiovascular Diseases prevention & control
- Abstract
The development of novel, non-invasive techniques and standardization of protocols to assess microvascular dysfunction have elucidated the key role of microvascular changes in the evolution of cardiovascular (CV) damage, and their capacity to predict an increased risk of adverse events. These technical advances parallel with the development of novel biological assays that enabled the ex vivo identification of pathways promoting microvascular dysfunction, providing novel potential treatment targets for preventing cerebral-CV disease. In this article, we provide an update of diagnostic testing strategies to detect and characterize microvascular dysfunction and suggestions on how to standardize and maximize the information obtained from each microvascular assay. We examine emerging data highlighting the significance of microvascular dysfunction in the development CV disease manifestations. Finally, we summarize the pathophysiology of microvascular dysfunction emphasizing the role of oxidative stress and its regulation by epigenetic mechanisms, which might represent potential targets for novel interventions beyond conventional approaches, representing a new frontier in CV disease reduction., (© The Author(s) 2020. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2021
- Full Text
- View/download PDF
37. Oxidative Stress and Hypertension.
- Author
-
Griendling KK, Camargo LL, Rios FJ, Alves-Lopes R, Montezano AC, and Touyz RM
- Subjects
- Animals, Antioxidants metabolism, Disease Models, Animal, Endoplasmic Reticulum metabolism, Humans, Hypertension metabolism, Inflammasomes physiology, Kidney metabolism, Mitochondria metabolism, NADPH Oxidases metabolism, Nitric Oxide metabolism, Nitric Oxide Synthase metabolism, Oxidation-Reduction, Signal Transduction physiology, Superoxides metabolism, Vascular Diseases metabolism, Hypertension etiology, Oxidative Stress physiology, Reactive Oxygen Species metabolism
- Abstract
A link between oxidative stress and hypertension has been firmly established in multiple animal models of hypertension but remains elusive in humans. While initial studies focused on inactivation of nitric oxide by superoxide, our understanding of relevant reactive oxygen species (superoxide, hydrogen peroxide, and peroxynitrite) and how they modify complex signaling pathways to promote hypertension has expanded significantly. In this review, we summarize recent advances in delineating the primary and secondary sources of reactive oxygen species (nicotinamide adenine dinucleotide phosphate oxidases, uncoupled endothelial nitric oxide synthase, endoplasmic reticulum, and mitochondria), the posttranslational oxidative modifications they induce on protein targets important for redox signaling, their interplay with endogenous antioxidant systems, and the role of inflammasome activation and endoplasmic reticular stress in the development of hypertension. We highlight how oxidative stress in different organ systems contributes to hypertension, describe new animal models that have clarified the importance of specific proteins, and discuss clinical studies that shed light on how these processes and pathways are altered in human hypertension. Finally, we focus on the promise of redox proteomics and systems biology to help us fully understand the relationship between ROS and hypertension and their potential for designing and evaluating novel antihypertensive therapies.
- Published
- 2021
- Full Text
- View/download PDF
38. Peripheral arteriopathy caused by Notch3 gain-of-function mutation involves ER and oxidative stress and blunting of NO/sGC/cGMP pathway.
- Author
-
Neves KB, Morris HE, Alves-Lopes R, Muir KW, Moreton F, Delles C, Montezano AC, and Touyz RM
- Subjects
- Animals, Arteries pathology, Brain metabolism, CADASIL genetics, CADASIL pathology, Cyclic GMP metabolism, Cytoplasmic Granules, Endoplasmic Reticulum Stress genetics, Endoplasmic Reticulum Stress physiology, Gain of Function Mutation, Humans, Mice, Mice, Transgenic, Muscle, Smooth, Vascular metabolism, Nitric Oxide metabolism, Oxidative Stress genetics, Oxidative Stress physiology, Signal Transduction, Soluble Guanylyl Cyclase, Vascular Diseases genetics, CADASIL metabolism, Muscle, Smooth, Vascular pathology, Receptor, Notch3 genetics, Vascular Diseases metabolism
- Abstract
Notch3 mutations cause Cerebral Autosomal Dominant Arteriopathy with Subcortical Infarcts and Leukoencephalopathy (CADASIL), which predisposes to stroke and dementia. CADASIL is characterised by vascular dysfunction and granular osmiophilic material (GOM) accumulation in cerebral small vessels. Systemic vessels may also be impacted by Notch3 mutations. However vascular characteristics and pathophysiological processes remain elusive. We investigated mechanisms underlying the peripheral vasculopathy mediated by CADASIL-causing Notch3 gain-of-function mutation. We studied: (i) small arteries and vascular smooth muscle cells (VSMCs) from TgNotch3R169C mice (CADASIL model), (ii) VSMCs from peripheral arteries from CADASIL patients, and (iii) post-mortem brains from CADASIL individuals. TgNotch3R169C vessels exhibited GOM deposits, increased vasoreactivity and impaired vasorelaxation. Hypercontractile responses were normalised by fasudil (Rho kinase inhibitor) and 4-phenylbutyrate (4-PBA; endoplasmic-reticulum (ER) stress inhibitor). Ca2+ transients and Ca2+ channel expression were increased in CADASIL VSMCs, with increased expression of Rho guanine nucleotide-exchange factors (GEFs) and ER stress proteins. Vasorelaxation mechanisms were impaired in CADASIL, evidenced by decreased endothelial nitric oxide synthase (eNOS) phosphorylation and reduced cyclic guanosine 3',5'-monophosphate (cGMP) levels, with associated increased soluble guanylate cyclase (sGC) oxidation, decreased sGC activity and reduced levels of the vasodilator hydrogen peroxide (H2O2). In VSMCs from CADASIL patients, sGC oxidation was increased and cGMP levels decreased, effects normalised by fasudil and 4-PBA. Cerebral vessels in CADASIL patients exhibited significant oxidative damage. In conclusion, peripheral vascular dysfunction in CADASIL is associated with altered Ca2+ homoeostasis, oxidative stress and blunted eNOS/sGC/cGMP signaling, processes involving Rho kinase and ER stress. We identify novel pathways underlying the peripheral arteriopathy induced by Notch3 gain-of-function mutation, phenomena that may also be important in cerebral vessels., (© 2021 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2021
- Full Text
- View/download PDF
39. ACE2/Ang-(1-7)/Mas1 axis and the vascular system: vasoprotection to COVID-19-associated vascular disease.
- Author
-
Kuriakose J, Montezano AC, and Touyz RM
- Subjects
- Animals, Blood Vessels enzymology, Humans, Proto-Oncogene Mas, Receptor, Angiotensin, Type 2 metabolism, Renin-Angiotensin System, SARS-CoV-2 metabolism, Vascular Diseases metabolism, Angiotensin I metabolism, Angiotensin-Converting Enzyme 2 metabolism, COVID-19 metabolism, Peptide Fragments metabolism, Proto-Oncogene Proteins metabolism, Receptors, G-Protein-Coupled metabolism, Vascular Diseases virology
- Abstract
The two axes of the renin-angiotensin system include the classical ACE/Ang II/AT1 axis and the counter-regulatory ACE2/Ang-(1-7)/Mas1 axis. ACE2 is a multifunctional monocarboxypeptidase responsible for generating Ang-(1-7) from Ang II. ACE2 is important in the vascular system where it is found in arterial and venous endothelial cells and arterial smooth muscle cells in many vascular beds. Among the best characterized functions of ACE2 is its role in regulating vascular tone. ACE2 through its effector peptide Ang-(1-7) and receptor Mas1 induces vasodilation and attenuates Ang II-induced vasoconstriction. In endothelial cells activation of the ACE2/Ang-(1-7)/Mas1 axis increases production of the vasodilator's nitric oxide and prostacyclin's and in vascular smooth muscle cells it inhibits pro-contractile and pro-inflammatory signaling. Endothelial ACE2 is cleaved by proteases, shed into the circulation and measured as soluble ACE2. Plasma ACE2 activity is increased in cardiovascular disease and may have prognostic significance in disease severity. In addition to its enzymatic function, ACE2 is the receptor for severe acute respiratory syndrome (SARS)-coronavirus (CoV) and SARS-Cov-2, which cause SARS and coronavirus disease-19 (COVID-19) respectively. ACE-2 is thus a double-edged sword: it promotes cardiovascular health while also facilitating the devastations caused by coronaviruses. COVID-19 is associated with cardiovascular disease as a risk factor and as a complication. Mechanisms linking COVID-19 and cardiovascular disease are unclear, but vascular ACE2 may be important. This review focuses on the vascular biology and (patho)physiology of ACE2 in cardiovascular health and disease and briefly discusses the role of vascular ACE2 as a potential mediator of vascular injury in COVID-19., (© 2021 The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
40. Importance of cholesterol-rich microdomains in the regulation of Nox isoforms and redox signaling in human vascular smooth muscle cells.
- Author
-
Anagnostopoulou A, Camargo LL, Rodrigues D, Montezano AC, and Touyz RM
- Subjects
- Animals, Cardiovascular Diseases pathology, Caveolin 1 genetics, Humans, Mice, Mice, Knockout, Muscle, Smooth, Vascular pathology, Oxidation-Reduction, Oxidative Stress, RNA, Small Interfering genetics, Signal Transduction, Cardiovascular Diseases metabolism, Caveolin 1 metabolism, Cholesterol metabolism, Membrane Microdomains metabolism, Muscle, Smooth, Vascular metabolism, NADPH Oxidase 1 metabolism, NADPH Oxidase 5 metabolism
- Abstract
Vascular smooth muscle cell (VSMC) function is regulated by Nox-derived reactive oxygen species (ROS) and redox-dependent signaling in discrete cellular compartments. Whether cholesterol-rich microdomains (lipid rafts/caveolae) are involved in these processes is unclear. Here we examined the sub-cellular compartmentalization of Nox isoforms in lipid rafts/caveolae and assessed the role of these microdomains in VSMC ROS production and pro-contractile and growth signaling. Intact small arteries and primary VSMCs from humans were studied. Vessels from Cav-1
-/- mice were used to test proof of concept. Human VSMCs express Nox1, Nox4, Nox5 and Cav-1. Cell fractionation studies showed that Nox1 and Nox5 but not Nox4, localize in cholesterol-rich fractions in VSMCs. Angiotensin II (Ang II) stimulation induced trafficking into and out of lipid rafts/caveolae for Nox1 and Nox5 respectively. Co-immunoprecipitation studies showed interactions between Cav-1/Nox1 but not Cav-1/Nox5. Lipid raft/caveolae disruptors (methyl-β-cyclodextrin (MCD) and Nystatin) and Ang II stimulation variably increased O2 - generation and phosphorylation of MLC20, Ezrin-Radixin-Moesin (ERM) and p53 but not ERK1/2, effects recapitulated in Cav-1 silenced (siRNA) VSMCs. Nox inhibition prevented Ang II-induced phosphorylation of signaling molecules, specifically, ERK1/2 phosphorylation was attenuated by mellitin (Nox5 inhibitor) and Nox5 siRNA, while p53 phosphorylation was inhibited by NoxA1ds (Nox1 inhibitor). Ang II increased oxidation of DJ1, dual anti-oxidant and signaling molecule, through lipid raft/caveolae-dependent processes. Vessels from Cav-1-/- mice exhibited increased O2 - generation and phosphorylation of ERM. We identify an important role for lipid rafts/caveolae that act as signaling platforms for Nox1 and Nox5 but not Nox4, in human VSMCs. Disruption of these microdomains promotes oxidative stress and Nox isoform-specific redox signalling important in vascular dysfunction associated with cardiovascular diseases.- Published
- 2020
- Full Text
- View/download PDF
41. Vascular toxicity associated with anti-angiogenic drugs.
- Author
-
Neves KB, Montezano AC, Lang NN, and Touyz RM
- Subjects
- Animals, Humans, Neoplasms blood supply, Neoplasms drug therapy, Receptors, Vascular Endothelial Growth Factor antagonists & inhibitors, Receptors, Vascular Endothelial Growth Factor metabolism, Signal Transduction, Vascular Endothelial Growth Factor A metabolism, Angiogenesis Inhibitors adverse effects, Angiogenesis Inhibitors therapeutic use, Neovascularization, Pathologic drug therapy
- Abstract
Over the past two decades, the treatment of cancer has been revolutionised by the highly successful introduction of novel molecular targeted therapies and immunotherapies, including small-molecule kinase inhibitors and monoclonal antibodies that target angiogenesis by inhibiting vascular endothelial growth factor (VEGF) signaling pathways. Despite their anti-angiogenic and anti-cancer benefits, the use of VEGF inhibitors (VEGFi) and other tyrosine kinase inhibitors (TKIs) has been hampered by potent vascular toxicities especially hypertension and thromboembolism. Molecular processes underlying VEGFi-induced vascular toxicities still remain unclear but inhibition of endothelial NO synthase (eNOS), reduced nitric oxide (NO) production, oxidative stress, activation of the endothelin system, and rarefaction have been implicated. However, the pathophysiological mechanisms still remain elusive and there is an urgent need to better understand exactly how anti-angiogenic drugs cause hypertension and other cardiovascular diseases (CVDs). This is especially important because VEGFi are increasingly being used in combination with other anti-cancer dugs, such as immunotherapies (immune checkpoint inhibitors (ICIs)), other TKIs, drugs that inhibit epigenetic processes (histone deacetylase (HDAC) inhibitor) and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, which may themselves induce cardiovascular injury. Here, we discuss vascular toxicities associated with TKIs, especially VEGFi, and provide an up-to-date overview on molecular mechanisms underlying VEGFi-induced vascular toxicity and cardiovascular sequelae. We also review the vascular effects of VEGFi when used in combination with other modern anti-cancer drugs., (© 2020 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2020
- Full Text
- View/download PDF
42. Comprehensive Characterization of the Vascular Effects of Cisplatin-Based Chemotherapy in Patients With Testicular Cancer.
- Author
-
Cameron AC, McMahon K, Hall M, Neves KB, Rios FJ, Montezano AC, Welsh P, Waterston A, White J, Mark PB, Touyz RM, and Lang NN
- Abstract
Background: Cisplatin-based chemotherapy increases the risk of cardiovascular and renal disease., Objectives: We aimed to define the time course, pathophysiology, and approaches to prevent cardiovascular disease associated with cisplatin-based chemotherapy., Methods: Two cohorts of patients with a history of testicular cancer (n = 53) were recruited. Cohort 1 consisted of 27 men undergoing treatment with: 1) surveillance; 2) 1 to 2 cycles of bleomycin, etoposide, and cisplatin (BEP) chemotherapy (low-intensity cisplatin); or 3) 3 to 4 cycles of BEP (high-intensity cisplatin). Endothelial function (percentage flow-mediated dilatation) and cardiovascular biomarkers were assessed at 6 visits over 9 months. Cohort 2 consisted of 26 men previously treated 1 to 7 years ago with surveillance or 3 to 4 cycles BEP. Vasomotor and fibrinolytic responses to bradykinin, acetylcholine, and sodium nitroprusside were evaluated using forearm venous occlusion plethysmography., Results: In cohort 1, the percentage flow-mediated dilatation decreased 24 h after the first cisplatin dose in patients managed with 3 to 4 cycles BEP (10.9 ± 0.9 vs. 16.7 ± 1.6; p < 0.01) but was unchanged from baseline thereafter. Six weeks after starting 3 to 4 cycles BEP, there were increased serum cholesterol levels (7.2 ± 0.5 mmol/l vs. 5.5 ± 0.2 mmol/l; p = 0.01), hemoglobin A1c (41.8 ± 2.0 mmol/l vs. 35.5 ± 1.2 mmol/l; p < 0.001), von Willebrand factor antigen (62.4 ± 5.4 mmol/l vs. 45.2 ± 2.8 mmol/l; p = 0.048) and cystatin C (0.91 ± 0.07 mmol/l vs. 0.65 ± 0.09 mmol/l; p < 0.01). In cohort 2, intra-arterial bradykinin, acetylcholine, and sodium nitroprusside caused dose-dependent vasodilation (p < 0.0001). Vasomotor responses, endogenous fibrinolytic factor release, and cardiovascular biomarkers were not different in patients managed with 3 to 4 cycles of BEP versus surveillance., Conclusions: Cisplatin-based chemotherapy induces acute and transient endothelial dysfunction, dyslipidemia, hyperglycemia, and nephrotoxicity in the early phases of treatment. Cardiovascular and renal protective strategies should target the early perichemotherapy period. (Clinical Characterisation of the Vascular Effects of Cis-platinum Based Chemotherapy in Patients With Testicular Cancer [VECTOR], NCT03557177; Intermediate and Long Term Vascular Effects of Cisplatin in Patients With Testicular Cancer [INTELLECT], NCT03557164)., (© 2020 The Authors.)
- Published
- 2020
- Full Text
- View/download PDF
43. Ca 2+ -Dependent NOX5 (NADPH Oxidase 5) Exaggerates Cardiac Hypertrophy Through Reactive Oxygen Species Production.
- Author
-
Zhao GJ, Zhao CL, Ouyang S, Deng KQ, Zhu L, Montezano AC, Zhang C, Hu F, Zhu XY, Tian S, Liu X, Ji YX, Zhang P, Zhang XJ, She ZG, Touyz RM, and Li H
- Subjects
- Angiotensin II pharmacology, Animals, Free Radical Scavengers pharmacology, Gene Expression Regulation drug effects, Humans, Isoenzymes metabolism, Mice, Mice, Transgenic, Myocytes, Cardiac metabolism, Oxidative Stress drug effects, Phagocytes enzymology, Rats, Signal Transduction drug effects, Vasoconstrictor Agents pharmacology, Ventricular Myosins metabolism, Acetylcysteine pharmacology, Cardiomegaly drug therapy, Cardiomegaly metabolism, Mitogen-Activated Protein Kinase Kinases metabolism, NADPH Oxidase 5 metabolism, Reactive Oxygen Species metabolism
- Abstract
NOX5 (NADPH oxidase 5) is a homolog of the gp91
phox subunit of the phagocyte NOX, which generates reactive oxygen species. NOX5 is involved in sperm motility and vascular contraction and has been implicated in diabetic nephropathy, atherosclerosis, and stroke. The function of NOX5 in the cardiac hypertrophy is unknown. Because NOX5 is a Ca2+ -sensitive, procontractile NOX isoform, we questioned whether it plays a role in cardiac hypertrophy. Studies were performed in (1) cardiac tissue from patients undergoing heart transplant for cardiomyopathy and heart failure, (2) NOX5-expressing rat cardiomyocytes, and (3) mice expressing human NOX5 in a cardiomyocyte-specific manner. Cardiac hypertrophy was induced in mice by transverse aorta coarctation and Ang II (angiotensin II) infusion. NOX5 expression was increased in human failing hearts. Rat cardiomyocytes infected with adenoviral vector encoding human NOX5 cDNA exhibited elevated reactive oxygen species levels with significant enlargement and associated increased expression of ANP (atrial natriuretic peptides) and β-MHC (β-myosin heavy chain) and prohypertrophic genes ( Nppa , Nppb , and Myh7 ) under Ang II stimulation. These effects were reduced by N-acetylcysteine and diltiazem. Pressure overload and Ang II infusion induced left ventricular hypertrophy, interstitial fibrosis, and contractile dysfunction, responses that were exaggerated in cardiac-specific NOX5 trangenic mice. These phenomena were associated with increased reactive oxygen species levels and activation of redox-sensitive MAPK (mitogen-activated protein kinase). N-acetylcysteine treatment reduced cardiac oxidative stress and attenuated cardiac hypertrophy in NOX5 trangenic. Our study defines Ca2+ -regulated NOX5 as an important NOX isoform involved in oxidative stress- and MAPK-mediated cardiac hypertrophy and contractile dysfunction.- Published
- 2020
- Full Text
- View/download PDF
44. Tissue sodium excess is not hypertonic and reflects extracellular volume expansion.
- Author
-
Rossitto G, Mary S, Chen JY, Boder P, Chew KS, Neves KB, Alves RL, Montezano AC, Welsh P, Petrie MC, Graham D, Touyz RM, and Delles C
- Subjects
- Aging metabolism, Animals, Edema diagnosis, Female, Humans, Hypertension diagnosis, Hypertension metabolism, Hypertension physiopathology, Liver metabolism, Lung metabolism, Male, Myocardium metabolism, Organ Specificity, Osmolar Concentration, Potassium metabolism, Rats, Inbred WKY, Skin metabolism, Transcription Factors metabolism, Edema metabolism, Homeostasis physiology, Sodium metabolism, Water-Electrolyte Balance physiology
- Abstract
Our understanding of Na
+ homeostasis has recently been reshaped by the notion of skin as a depot for Na+ accumulation in multiple cardiovascular diseases and risk factors. The proposed water-independent nature of tissue Na+ could induce local pathogenic changes, but lacks firm demonstration. Here, we show that tissue Na+ excess upon high Na+ intake is a systemic, rather than skin-specific, phenomenon reflecting architectural changes, i.e. a shift in the extracellular-to-intracellular compartments, due to a reduction of the intracellular or accumulation of water-paralleled Na+ in the extracellular space. We also demonstrate that this accumulation is unlikely to justify the observed development of experimental hypertension if it were water-independent. Finally, we show that this isotonic skin Na+ excess, reflecting subclinical oedema, occurs in hypertensive patients and in association with aging. The implications of our findings, questioning previous assumptions but also reinforcing the importance of tissue Na+ excess, are both mechanistic and clinical.- Published
- 2020
- Full Text
- View/download PDF
45. Epidermal growth factor signaling through transient receptor potential melastatin 7 cation channel regulates vascular smooth muscle cell function.
- Author
-
Zou ZG, Rios FJ, Neves KB, Alves-Lopes R, Ling J, Baillie GS, Gao X, Fuller W, Camargo LL, Gudermann T, Chubanov V, Montezano AC, and Touyz RM
- Subjects
- Animals, CSK Tyrosine-Protein Kinase metabolism, Calcium metabolism, Cation Transport Proteins metabolism, Extracellular Signal-Regulated MAP Kinases metabolism, HEK293 Cells, Humans, Magnesium metabolism, Mice, Inbred C57BL, Morphogenesis, Muscle, Smooth, Vascular growth & development, Phosphorylation, Primary Cell Culture, Rats, Inbred WKY, Epidermal Growth Factor metabolism, Muscle, Smooth, Vascular metabolism, Myocytes, Smooth Muscle metabolism, Protein Serine-Threonine Kinases metabolism, TRPM Cation Channels metabolism
- Abstract
Objective: Transient receptor potential (TRP) melastatin 7 (TRPM7) cation channel, a dual-function ion channel/protein kinase, regulates vascular smooth muscle cell (VSMC) Mg2+ homeostasis and mitogenic signaling. Mechanisms regulating vascular growth effects of TRPM7 are unclear, but epidermal growth factor (EGF) may be important because it is a magnesiotropic hormone involved in cellular Mg2+ regulation and VSMC proliferation. Here we sought to determine whether TRPM7 is a downstream target of EGF in VSMCs and if EGF receptor (EGFR) through TRPM7 influences VSMC function. Approach and results: Studies were performed in primary culture VSMCs from rats and humans and vascular tissue from mice deficient in TRPM7 (TRPM7+/Δkinase and TRPM7R/R). EGF increased expression and phosphorylation of TRPM7 and stimulated Mg2+ influx in VSMCs, responses that were attenuated by gefitinib (EGFR inhibitor) and NS8593 (TRPM7 inhibitor). Co-immunoprecipitation (IP) studies, proximity ligation assay (PLA) and live-cell imaging demonstrated interaction of EGFR and TRPM7, which was enhanced by EGF. PP2 (c-Src inhibitor) decreased EGF-induced TRPM7 activation and prevented EGFR-TRPM7 association. EGF-stimulated migration and proliferation of VSMCs were inhibited by gefitinib, PP2, NS8593 and PD98059 (ERK1/2 inhibitor). Phosphorylation of EGFR and ERK1/2 was reduced in VSMCs from TRPM7+/Δkinase mice, which exhibited reduced aortic wall thickness and decreased expression of PCNA and Notch 3, findings recapitulated in TRPM7R/R mice., Conclusions: We show that EGFR directly interacts with TRPM7 through c-Src-dependent processes. Functionally these phenomena regulate [Mg2+]i homeostasis, ERK1/2 signaling and VSMC function. Our findings define a novel signaling cascade linking EGF/EGFR and TRPM7, important in vascular homeostasis., (© 2020 The Author(s).)
- Published
- 2020
- Full Text
- View/download PDF
46. Selective ETA vs. dual ETA/B receptor blockade for the prevention of sunitinib-induced hypertension and albuminuria in WKY rats.
- Author
-
Mirabito Colafella KM, Neves KB, Montezano AC, Garrelds IM, van Veghel R, de Vries R, Uijl E, Baelde HJ, van den Meiracker AH, Touyz RM, Danser AHJ, and Versmissen J
- Subjects
- Albuminuria chemically induced, Albuminuria metabolism, Albuminuria pathology, Animals, Arteries metabolism, Arteries physiopathology, Disease Models, Animal, Epoprostenol metabolism, Hypertension chemically induced, Hypertension metabolism, Hypertension physiopathology, Isoxazoles pharmacology, Kidney drug effects, Kidney metabolism, Kidney physiopathology, Male, Oxidative Stress drug effects, Pyrimidines pharmacology, Rats, Inbred WKY, Receptor, Endothelin A drug effects, Receptor, Endothelin A metabolism, Receptor, Endothelin B drug effects, Receptor, Endothelin B metabolism, Signal Transduction, Sulfonamides pharmacology, Sunitinib, Thiophenes pharmacology, Albuminuria prevention & control, Antihypertensive Agents pharmacology, Arteries drug effects, Blood Pressure drug effects, Endothelin A Receptor Antagonists pharmacology, Endothelin B Receptor Antagonists pharmacology, Hypertension prevention & control
- Abstract
Aims: Although effective in preventing tumour growth, angiogenesis inhibitors cause off-target effects including cardiovascular toxicity and renal injury, most likely via endothelin (ET)-1 up-regulation. ET-1 via stimulation of the ETA receptor has pro-hypertensive actions whereas stimulation of the ETB receptor can elicit both pro- or anti-hypertensive effects. In this study, our aim was to determine the efficacy of selective ETA vs. dual ETA/B receptor blockade for the prevention of angiogenesis inhibitor-induced hypertension and albuminuria., Methods and Results: Male Wistar Kyoto (WKY) rats were treated with vehicle, sunitinib (angiogenesis inhibitor; 14 mg/kg/day) alone or in combination with macitentan (ETA/B receptor antagonist; 30 mg/kg/day) or sitaxentan (selective ETA receptor antagonist; 30 or 100 mg/kg/day) for 8 days. Compared with vehicle, sunitinib treatment caused a rapid and sustained increase in mean arterial pressure of ∼25 mmHg. Co-treatment with macitentan or sitaxentan abolished the pressor response to sunitinib. Sunitinib did not induce endothelial dysfunction. However, it was associated with increased aortic, mesenteric, and renal oxidative stress, an effect that was absent in mesenteric arteries of the macitentan and sitaxentan co-treated groups. Albuminuria was greater in the sunitinib- than vehicle-treated group. Co-treatment with sitaxentan, but not macitentan, prevented this increase in albuminuria. Sunitinib treatment increased circulating and urinary prostacyclin levels and had no effect on thromboxane levels. These increases in prostacyclin were blunted by co-treatment with sitaxentan., Conclusions: Our results demonstrate that both selective ETA and dual ETA/B receptor antagonism prevents sunitinib-induced hypertension, whereas sunitinib-induced albuminuria was only prevented by selective ETA receptor antagonism. In addition, our results uncover a role for prostacyclin in the development of these effects. In conclusion, selective ETA receptor antagonism is sufficient for the prevention of sunitinib-induced hypertension and renal injury., (Published on behalf of the European Society of Cardiology. All rights reserved. © The Author(s) 2019. For permissions, please email: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
47. Oxidative Stress: A Unifying Paradigm in Hypertension.
- Author
-
Touyz RM, Rios FJ, Alves-Lopes R, Neves KB, Camargo LL, and Montezano AC
- Subjects
- Aldosterone metabolism, Angiotensin II metabolism, Endothelin-1 metabolism, Endothelium, Vascular physiopathology, Humans, Inflammation physiopathology, Intercellular Signaling Peptides and Proteins metabolism, Oxidation-Reduction, Reactive Oxygen Species metabolism, Sex Factors, Signal Transduction physiology, Vascular Remodeling physiology, Vascular Stiffness physiology, Vasoconstriction physiology, Hypertension physiopathology, Oxidative Stress physiology
- Abstract
The etiology of hypertension involves complex interactions among genetic, environmental, and pathophysiologic factors that influence many regulatory systems. Hypertension is characteristically associated with vascular dysfunction, cardiovascular remodelling, renal dysfunction, and stimulation of the sympathetic nervous system. Emerging evidence indicates that the immune system is also important and that activated immune cells migrate and accumulate in tissues promoting inflammation, fibrosis, and target-organ damage. Common to these processes is oxidative stress, defined as an imbalance between oxidants and antioxidants in favour of the oxidants that leads to a disruption of oxidation-reduction (redox) signalling and control and molecular damage. Physiologically, reactive oxygen species (ROS) act as signalling molecules and influence cell function through highly regulated redox-sensitive signal transduction. In hypertension, oxidative stress promotes posttranslational modification (oxidation and phosphorylation) of proteins and aberrant signalling with consequent cell and tissue damage. Many enzymatic systems generate ROS, but NADPH oxidases (Nox) are the major sources in cells of the heart, vessels, kidneys, and immune system. Expression and activity of Nox are increased in hypertension and are the major systems responsible for oxidative stress in cardiovascular disease. Here we provide a unifying concept where oxidative stress is a common mediator underlying pathophysiologic processes in hypertension. We focus on some novel concepts whereby ROS influence vascular function, aldosterone/mineralocorticoid actions, and immunoinflammation, all important processes contributing to the development of hypertension., (Copyright © 2020 The Authors. Published by Elsevier Inc. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
48. Local endothelial DNA repair deficiency causes aging-resembling endothelial-specific dysfunction.
- Author
-
Bautista-Niño PK, Portilla-Fernandez E, Rubio-Beltrán E, van der Linden JJ, de Vries R, van Veghel R, de Boer M, Durik M, Ridwan Y, Brandt R, Essers J, Menzies RI, Thomas R, de Bruin A, Duncker DJ, van Beusekom HMM, Ghanbari M, Hoeijmakers JHJ, Sedlacek R, Touyz RM, Montezano AC, van der Pluijm I, Danser AHJ, Haanes KA, and Roks AJM
- Subjects
- Age Factors, Aging metabolism, Aging pathology, Animals, Capillary Permeability, Cyclin-Dependent Kinase Inhibitor p21 genetics, Cyclin-Dependent Kinase Inhibitor p21 metabolism, DNA-Binding Proteins genetics, Endonucleases genetics, Endothelial Cells pathology, Endothelium, Vascular pathology, Endothelium, Vascular physiopathology, Mice, Inbred C57BL, Mice, Knockout, Nitric Oxide metabolism, Nitric Oxide Synthase Type III metabolism, Superoxides metabolism, Vascular Stiffness, Vasodilation, Aging genetics, Cellular Senescence genetics, DNA Damage, DNA Repair, DNA-Binding Proteins deficiency, Endonucleases deficiency, Endothelial Cells metabolism, Endothelium, Vascular metabolism
- Abstract
We previously identified genomic instability as a causative factor for vascular aging. In the present study, we determined which vascular aging outcomes are due to local endothelial DNA damage, which was accomplished by genetic removal of ERCC1 (excision repair cross-complementation group 1) DNA repair in mice (EC-knockout (EC-KO) mice). EC-KO showed a progressive decrease in microvascular dilation of the skin, increased microvascular leakage in the kidney, decreased lung perfusion, and increased aortic stiffness compared with wild-type (WT). EC-KO showed expression of DNA damage and potential senescence marker p21 exclusively in the endothelium, as demonstrated in aorta. Also the kidney showed p21-positive cells. Vasodilator responses measured in organ baths were decreased in aorta, iliac and coronary artery EC-KO compared with WT, of which coronary artery was the earliest to be affected. Nitric oxide-mediated endothelium-dependent vasodilation was abolished in aorta and coronary artery, whereas endothelium-derived hyperpolarization and responses to exogenous nitric oxide (NO) were intact. EC-KO showed increased superoxide production compared with WT, as measured in lung tissue, rich in endothelial cells (ECs). Arterial systolic blood pressure (BP) was increased at 3 months, but normal at 5 months, at which age cardiac output (CO) was decreased. Since no further signs of cardiac dysfunction were detected, this decrease might be an adaptation to prevent an increase in BP. In summary, a selective DNA repair defect in the endothelium produces features of age-related endothelial dysfunction, largely attributed to loss of endothelium-derived NO. Increased superoxide generation might contribute to the observed changes affecting end organ perfusion, as demonstrated in kidney and lung., (© 2020 The Author(s). Published by Portland Press Limited on behalf of the Biochemical Society.)
- Published
- 2020
- Full Text
- View/download PDF
49. Lessons Learned From RAG-1-Deficient Mice in Hypertension.
- Author
-
Rios FJ, Montezano AC, and Touyz RM
- Subjects
- Animals, Homeodomain Proteins genetics, Laboratories, Mice, Mice, Knockout, Phenotype, Angiotensin II, Hypertension genetics
- Published
- 2020
- Full Text
- View/download PDF
50. Chanzyme TRPM7 protects against cardiovascular inflammation and fibrosis.
- Author
-
Rios FJ, Zou ZG, Harvey AP, Harvey KY, Nosalski R, Anyfanti P, Camargo LL, Lacchini S, Ryazanov AG, Ryazanova L, McGrath S, Guzik TJ, Goodyear CS, Montezano AC, and Touyz RM
- Subjects
- Animals, Cardiomegaly genetics, Cardiomegaly pathology, Cardiomegaly physiopathology, Cardiomyopathies genetics, Cardiomyopathies pathology, Cardiomyopathies physiopathology, Cell Proliferation, Cells, Cultured, Coculture Techniques, Fibroblasts metabolism, Fibroblasts pathology, Fibrosis, Inflammation genetics, Inflammation pathology, Inflammation physiopathology, Leukocyte Rolling, Macrophages metabolism, Macrophages pathology, Magnesium metabolism, Male, Mice, 129 Strain, Mice, Inbred C57BL, Mice, Knockout, Myocardium pathology, Signal Transduction, TRPM Cation Channels deficiency, TRPM Cation Channels genetics, Transendothelial and Transepithelial Migration, Cardiomegaly metabolism, Cardiomyopathies metabolism, Inflammation metabolism, Inflammation Mediators metabolism, Myocardium metabolism, TRPM Cation Channels metabolism, Ventricular Remodeling
- Abstract
Aims: Transient Receptor Potential Melastatin 7 (TRPM7) cation channel is a chanzyme (channel + kinase) that influences cellular Mg2+ homeostasis and vascular signalling. However, the pathophysiological significance of TRPM7 in the cardiovascular system is unclear. The aim of this study was to investigate the role of this chanzyme in the cardiovascular system focusing on inflammation and fibrosis., Methods and Results: TRPM7-deficient mice with deletion of the kinase domain (TRPM7+/Δkinase) were studied and molecular mechanisms investigated in TRPM7+/Δkinase bone marrow-derived macrophages (BMDM) and co-culture systems with cardiac fibroblasts. TRPM7-deficient mice had significant cardiac hypertrophy, fibrosis, and inflammation. Cardiac collagen and fibronectin content, expression of pro-inflammatory mediators (SMAD3, TGFβ) and cytokines [interleukin (IL)-6, IL-10, IL-12, tumour necrosis factor-α] and phosphorylation of the pro-inflammatory signalling molecule Stat1, were increased in TRPM7+/Δkinase mice. These processes were associated with infiltration of inflammatory cells (F4/80+CD206+ cardiac macrophages) and increased galectin-3 expression. Cardiac [Mg2+]i, but not [Ca2+]i, was reduced in TRPM7+/Δkinase mice. Calpain, a downstream TRPM7 target, was upregulated (increased expression and activation) in TRPM7+/Δkinase hearts. Vascular functional and inflammatory responses, assessed in vivo by intra-vital microscopy, demonstrated impaired neutrophil rolling, increased neutrophil: endothelial attachment and transmigration of leucocytes in TRPM7+/Δkinase mice. TRPM7+/Δkinase BMDMs had increased levels of galectin-3, IL-10, and IL-6. In co-culture systems, TRPM7+/Δkinase macrophages increased expression of fibronectin, proliferating cell nuclear antigen, and TGFβ in cardiac fibroblasts from wild-type mice, effects ameliorated by MgCl2 treatment., Conclusions: We identify a novel anti-inflammatory and anti-fibrotic role for TRPM7 and suggest that its protective effects are mediated, in part, through Mg2+-sensitive processes., (© The Author(s) 2019. Published by Oxford University Press on behalf of the European Society of Cardiology.)
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.