29 results on '"Francisco J. Rios"'
Search Results
2. Editorial: Receptors in cardiovascular diseases: Mechanisms, diagnosis, and treatment
- Author
-
Shutong Shen, Francisco J. Rios, and Phung N. Thai
- Subjects
cardiovascular disease ,receptors ,GPCR (G protein coupled receptors) ,myocardial infarction ,cardiovascular toxicity ,cardiac fibrosis ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Published
- 2023
- Full Text
- View/download PDF
3. Cardiovascular toxicity of tyrosine kinase inhibitors during cancer treatment: Potential involvement of TRPM7
- Author
-
Qing Liu, Suyao Li, Yuran Qiu, Jiayu Zhang, Francisco J. Rios, Zhiguo Zou, and Rhian M. Touyz
- Subjects
receptor tyrosine kinase ,TRPM7 ,cardiovascular toxicities ,cancer ,tyrosine kinase inhibitors ,magnesium ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Receptor tyrosine kinases (RTKs) are a class of membrane spanning cell-surface receptors that transmit extracellular signals through the membrane to trigger diverse intracellular signaling through tyrosine kinases (TKs), and play important role in cancer development. Therapeutic approaches targeting RTKs such as vascular endothelial growth factor receptor (VEGFR), epidermal growth factor receptor (EGFR), and platelet-derived growth factor receptor (PDGFR), and TKs, such as c-Src, ABL, JAK, are widely used to treat human cancers. Despite favorable benefits in cancer treatment that prolong survival, these tyrosine kinase inhibitors (TKIs) and monoclonal antibodies targeting RTKs are also accompanied by adverse effects, including cardiovascular toxicity. Mechanisms underlying TKI-induced cardiovascular toxicity remain unclear. The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed chanzyme consisting of a membrane-based ion channel and intracellular α-kinase. TRPM7 is a cation channel that regulates transmembrane Mg2+ and Ca2+ and is involved in a variety of (patho)physiological processes in the cardiovascular system, contributing to hypertension, cardiac fibrosis, inflammation, and atrial arrhythmias. Of importance, we and others demonstrated significant cross-talk between TRPM7, RTKs, and TK signaling in different cell types including vascular smooth muscle cells (VSMCs), which might be a link between TKIs and their cardiovascular effects. In this review, we summarize the implications of RTK inhibitors (RTKIs) and TKIs in cardiovascular toxicities during anti-cancer treatment, with a focus on the potential role of TRPM7/Mg2+ as a mediator of RTKI/TKI-induced cardiovascular toxicity. We also describe the important role of TRPM7 in cancer development and cardiovascular diseases, and the interaction between TRPM7 and RTKs, providing insights for possible mechanisms underlying cardiovascular disease in cancer patients treated with RTKI/TKIs.
- Published
- 2023
- Full Text
- View/download PDF
4. Comprehensive Characterization of the Vascular Effects of Cisplatin-Based Chemotherapy in Patients With Testicular Cancer
- Author
-
Alan C. Cameron, MBChB, PhD, Kelly McMahon, BSc, Mark Hall, MBChB, Karla B. Neves, PhD, Francisco J. Rios, PhD, Augusto C. Montezano, PhD, Paul Welsh, PhD, Ashita Waterston, MBChB, PhD, Jeff White, MBChB, DM, Patrick B. Mark, MBChB, PhD, Rhian M. Touyz, MBBCh, PhD, and Ninian N. Lang, MBChB, PhD
- Subjects
germ cell tumors ,platinum therapy ,testicular cancer ,thrombosis ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - 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)
- Published
- 2020
- Full Text
- View/download PDF
5. NADPH Oxidase 5 Is a Pro‐Contractile Nox Isoform and a Point of Cross‐Talk for Calcium and Redox Signaling‐Implications in Vascular Function
- Author
-
Augusto C. Montezano, Livia De Lucca Camargo, Patrik Persson, Francisco J. Rios, Adam P. Harvey, Aikaterini Anagnostopoulou, Roberto Palacios, Ana Caroline P. Gandara, Rheure Alves‐Lopes, Karla B. Neves, Maria Dulak‐Lis, Chet E. Holterman, Pedro Lagerblad de Oliveira, Delyth Graham, Christopher Kennedy, and Rhian M. Touyz
- Subjects
cell signaling ,contraction ,vascular biology ,Diseases of the circulatory (Cardiovascular) system ,RC666-701 - Abstract
Background NADPH Oxidase 5 (Nox5) is a calcium‐sensitive superoxide‐generating Nox. It is present in lower forms and higher mammals, but not in rodents. Nox5 is expressed in vascular cells, but the functional significance remains elusive. Given that contraction is controlled by calcium and reactive oxygen species, both associated with Nox5, we questioned the role of Nox5 in pro‐contractile signaling and vascular function. Methods and Results Transgenic mice expressing human Nox5 in a vascular smooth muscle cell–specific manner (Nox5 mice) and Rhodnius prolixus, an arthropod model that expresses Nox5 endogenoulsy, were studied. Reactive oxygen species generation was increased systemically and in the vasculature and heart in Nox5 mice. In Nox5‐expressing mice, agonist‐induced vasoconstriction was exaggerated and endothelium‐dependent vasorelaxation was impaired. Vascular structural and mechanical properties were not influenced by Nox5. Vascular contractile responses in Nox5 mice were normalized by N‐acetylcysteine and inhibitors of calcium channels, calmodulin, and endoplasmic reticulum ryanodine receptors, but not by GKT137831 (Nox1/4 inhibitor). At the cellular level, vascular changes in Nox5 mice were associated with increased vascular smooth muscle cell [Ca2+]i, increased reactive oxygen species and nitrotyrosine levels, and hyperphosphorylation of pro‐contractile signaling molecules MLC20 (myosin light chain 20) and MYPT1 (myosin phosphatase target subunit 1). Blood pressure was similar in wild‐type and Nox5 mice. Nox5 did not amplify angiotensin II effects. In R. prolixus, gastrointestinal smooth muscle contraction was blunted by Nox5 silencing, but not by VAS2870 (Nox1/2/4 inhibitor). Conclusions Nox5 is a pro‐contractile Nox isoform important in redox‐sensitive contraction. This involves calcium‐calmodulin and endoplasmic reticulum–regulated mechanisms. Our findings define a novel function for vascular Nox5, linking calcium and reactive oxygen species to the pro‐contractile molecular machinery in vascular smooth muscle cells.
- Published
- 2018
- Full Text
- View/download PDF
6. Interplay between Hormones, the Immune System, and Metabolic Disorders
- Author
-
Francisco J. Rios, Naïma Moustaïd-Moussa, and Joilson O. Martins
- Subjects
Pathology ,RB1-214 - Published
- 2018
- Full Text
- View/download PDF
7. TRPM7, Magnesium, and Signaling
- Author
-
Zhi-Guo Zou, Francisco J. Rios, Augusto C. Montezano, and Rhian M. Touyz
- Subjects
TRPM7 ,magnesium transporters ,receptor tyrosine kinases ,VEGFR ,EGFR ,Biology (General) ,QH301-705.5 ,Chemistry ,QD1-999 - Abstract
The transient receptor potential melastatin-subfamily member 7 (TRPM7) is a ubiquitously expressed chanzyme that possesses an ion channel permeable to the divalent cations Mg2+, Ca2+, and Zn2+, and an α-kinase that phosphorylates downstream substrates. TRPM7 and its homologue TRPM6 have been implicated in a variety of cellular functions and is critically associated with intracellular signaling, including receptor tyrosine kinase (RTK)-mediated pathways. Emerging evidence indicates that growth factors, such as EGF and VEGF, signal through their RTKs, which regulate activity of TRPM6 and TRPM7. TRPM6 is primarily an epithelial-associated channel, while TRPM7 is more ubiquitous. In this review we focus on TRPM7 and its association with growth factors, RTKs, and downstream kinase signaling. We also highlight how interplay between TRPM7, Mg2+ and signaling kinases influences cell function in physiological and pathological conditions, such as cancer and preeclampsia.
- Published
- 2019
- Full Text
- View/download PDF
8. (+)-(S)-N-[(1-Benzothiophen-2-yl)methylidene]-1-(naphthalen-1-yl)ethylamine
- Author
-
Guadalupe Hernández-Téllez, Oscar Portillo-Moreno, René Gutiérrez, Francisco J. Rios-Merino, and Angel Mendoza
- Subjects
Crystallography ,QD901-999 - Abstract
In the title compound, C21H17NS, the C=N double bond shows an E conformation. The dihedral angle between the mean planes of the naphthyl residue and the benzothiophene residue is 89.14 (6)°. The crystal packing is stabilized by intermolecular C—H...π interactions, building a ribbon structure along the a axis.
- Published
- 2013
- Full Text
- View/download PDF
9. Oxidized LDL Induces Alternative Macrophage Phenotype through Activation of CD36 and PAFR
- Author
-
Francisco J. Rios, Marianna M. Koga, Mateus Pecenin, Matheus Ferracini, Magnus Gidlund, and S. Jancar
- Subjects
Pathology ,RB1-214 - Abstract
OxLDL is recognized by macrophage scavenger receptors, including CD36; we have recently found that Platelet-Activating Factor Receptor (PAFR) is also involved. Since PAFR in macrophages is associated with suppressor function, we examined the effect of oxLDL on macrophage phenotype. It was found that the presence of oxLDL during macrophage differentiation induced high mRNA levels to IL-10, mannose receptor, PPARγ and arginase-1 and low levels of IL-12 and iNOS. When human THP-1 macrophages were pre-treated with oxLDL then stimulated with LPS, the production of IL-10 and TGF-β significantly increased, whereas that of IL-6 and IL-8 decreased. In murine TG-elicited macrophages, this protocol significantly reduced NO, iNOS and COX2 expression. Thus, oxLDL induced macrophage differentiation and activation towards the alternatively activated M2-phenotype. In murine macrophages, oxLDL induced TGF-β, arginase-1 and IL-10 mRNA expression, which were significantly reduced by pre-treatment with PAFR antagonists (WEB and CV) or with antibodies to CD36. The mRNA expression of IL-12, RANTES and CXCL2 were not affected. We showed that this profile of macrophage activation is dependent on the engagement of both CD36 and PAFR. We conclude that oxLDL induces alternative macrophage activation by mechanisms involving CD36 and PAFR.
- Published
- 2013
- Full Text
- View/download PDF
10. Expression of PAFR as Part of a Prosurvival Response to Chemotherapy: A Novel Target for Combination Therapy in Melanoma
- Author
-
Ana Claudia Onuchic, Camila M. L. Machado, Renata F. Saito, Francisco J. Rios, Sônia Jancar, and Roger Chammas
- Subjects
Pathology ,RB1-214 - Abstract
Melanoma cells express the platelet-activating factor receptor (PAFR) and, thus, respond to PAF, a bioactive lipid produced by both tumour cells and those in the tumour microenvironment such as macrophages. Here, we show that treatment of a human melanoma SKmel37 cell line with cisplatin led to increased expression of PAFR and its accumulation. In the presence of exogenous PAF, melanoma cells were significantly more resistant to cisplatin-induced cell death. Inhibition of PAFR-dependent signalling pathways by a PAFR antagonist (WEB2086) showed chemosensitisation of melanoma cells in vitro. Nude mice were inoculated with SKmel37 cells and treated with cisplatin and WEB2086. Animals treated with both agents showed significantly decreased tumour growth compared to the control group and groups treated with only one agent. PAFR accumulation and signalling are part of a prosurvival program of melanoma cells, therefore constituting a promising target for combination therapy for melanomas.
- Published
- 2012
- Full Text
- View/download PDF
11. Comprehensive Characterization of the Vascular Effects of Cisplatin-Based Chemotherapy in Patients With Testicular Cancer
- Author
-
Paul Welsh, Ashita Waterston, Augusto C. Montezano, Francisco J. Rios, Jeff White, Karla B Neves, Rhian M. Touyz, Patrick B. Mark, Kelly R. McMahon, Alan C. Cameron, Mark Hall, and Ninian N. Lang
- Subjects
Oncology ,lcsh:Diseases of the circulatory (Cardiovascular) system ,medicine.medical_specialty ,medicine.medical_treatment ,cisplatin ,BEP, bleomycin, etoposide and cisplatin ,vWF, von Willebrand factor ,atherosclerosis, cancer ,chemotherapy ,Bleomycin ,lcsh:RC254-282 ,endothelial dysfunction ,Nephrotoxicity ,ICAM, intracellular adhesion molecule ,chemistry.chemical_compound ,Internal medicine ,platinum therapy ,medicine ,0FMD, flow-mediated dilatation ,Endothelial dysfunction ,germ cell tumors ,t-PA, tissue plasminogen activator ,Testicular cancer ,Etoposide ,thrombosis ,Original Research ,Cisplatin ,Chemotherapy ,business.industry ,ACh, acetylcholine ,lcsh:Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,medicine.disease ,testicular cancer ,chemistry ,lcsh:RC666-701 ,Cohort ,SNP, sodium nitroprusside ,PAI, plasminogen activator inhibitor ,vasoreactivity ,FBF, forearm blood flow ,Cardiology and Cardiovascular Medicine ,business ,BK, bradykinin ,Editorial Comment ,medicine.drug - 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), Central Illustration
- Published
- 2020
12. Epidermal growth factor signaling through transient receptor potential melastatin 7 cation channel regulates vascular smooth muscle cell function
- Author
-
Livia L Camargo, Augusto C. Montezano, William Fuller, Rheure Alves-Lopes, Vladimir Chubanov, Thomas Gudermann, ZhiGuo Zou, Xing Gao, Rhian M. Touyz, George S. Baillie, Francisco J. Rios, Jiayue Ling, and Karla B Neves
- Subjects
Vascular smooth muscle ,Myocytes, Smooth Muscle ,Primary Cell Culture ,TRPM7 ,TRPM Cation Channels ,Protein Serine-Threonine Kinases ,Rats, Inbred WKY ,Molecular Bases of Health & Disease ,Muscle, Smooth, Vascular ,CSK Tyrosine-Protein Kinase ,Gefitinib ,Epidermal growth factor ,medicine ,Morphogenesis ,Animals ,Humans ,Magnesium ,Phosphorylation ,Protein kinase A ,Receptor ,Extracellular Signal-Regulated MAP Kinases ,Cation Transport Proteins ,Research Articles ,EGFR inhibitors ,EGF ,Epidermal Growth Factor ,Chemistry ,General Medicine ,Signaling ,Cell biology ,Mice, Inbred C57BL ,HEK293 Cells ,Cardiovascular System & Vascular Biology ,vascular smooth muscle ,Calcium ,medicine.drug - 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.
- Published
- 2020
13. Central role of c-Src in NOX5- mediated redox signalling in vascular smooth muscle cells in human hypertension
- Author
-
Misbah Hussain, ZhiGuo Zou, Francisco J. Rios, Fazli Rabbi Awan, Yu Wang, Augusto C. Montezano, Rhian M. Touyz, Thomas Jensen, Rheure Alves-Lopes, Tomasz J. Guzik, Richard C. Hartley, Karla B Neves, and Livia L Camargo
- Subjects
0301 basic medicine ,Cell signaling ,Vascular smooth muscle ,Physiology ,Myocytes, Smooth Muscle ,Hyperphosphorylation ,Mice, Transgenic ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Muscle, Smooth, Vascular ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,medicine ,Animals ,Humans ,RNA, Small Interfering ,Cells, Cultured ,Protein kinase C ,biology ,Chemistry ,Angiotensin II ,Protein-Tyrosine Kinases ,Melitten ,Actins ,Cell biology ,030104 developmental biology ,NADPH Oxidase 5 ,Hypertension ,cardiovascular system ,biology.protein ,Phosphorylation ,P22phox ,Signal transduction ,Reactive Oxygen Species ,Cardiology and Cardiovascular Medicine ,Oxidation-Reduction ,Oxidative stress - Abstract
AIMS NOX-derived reactive oxygen species (ROS) are mediators of signaling 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 signaling molecules, 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 downregulated 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 upregulated, with associated oxidative stress, hyperoxidation (c-Src, peroxiredoxin, DJ-1) and hyperphosphorylation (PKC, ERK1/2, MLC20) of signaling 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. CONCLUSIONS We define NOX5/ROS/c-Src as a novel feedforward signaling 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. TRANSLATIONAL PERSPECTIVE Oxidative stress is a major factor contributing to vascular damage in hypertension. We corroborate experimental evidence that NOX-derived ROS generation is increased in human vascular smooth muscle cells (VSMC) and demonstrate that in human hypertension NOX5 upregulation is a major trigger of VSMC dysfunction. We uncover new regulatory molecular mechanisms of NOX5 and define NOX5/ROS/c-Src as a novel signaling pathway in human VSMCs. This system is augmented in hypertension contributing to abnormal VSMC redox signaling, cytoskeletal disorganization and vascular dysfunction. Modulating the NOX5/ROS/c-Src pathway may have therapeutic potential by targeting redox signaling pathways involved in vascular dysfunction associated with hypertension.
- Published
- 2022
14. Cardiovascular and renal risk factors and complications associated with COVID-19
- Author
-
Augusto C. Montezano, John R. Petrie, Marcus O.E. Boyd, Tomasz J. Guzik, Francisco J. Rios, Rhian M. Touyz, Patrick B. Mark, Sandosh Padmanabhan, Linsay McCallum, Robert Sykes, Christian Delles, and Colin Berry
- Subjects
cardiovascular risk ,long COVID-19 ,hypertension ,diabetes ,business.industry ,SARS-CoV-2 ,kidney disease ,Acute kidney injury ,ACE2 ,Context (language use) ,Disease ,Review ,medicine.disease ,Bioinformatics ,Respiratory failure ,Diabetes mellitus ,Pathophysiology of hypertension ,RC666-701 ,medicine ,myocarditis thromboembolic disease ,Diseases of the circulatory (Cardiovascular) system ,Cardiology and Cardiovascular Medicine ,business ,Dyslipidemia ,Kidney disease - 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). Résumé: La pandémie actuelle de COVID-19 causée par le coronavirus du syndrome respiratoire aigu sévère 2 (SRAS-CoV-2) est le plus grand enjeu médical des dernières décennies. Elle a mis en évidence des vulnérabilités cardiovasculaires imprévues à tous les stades de la COVID-19 (avant l'infection, pendant la phase aiguë et pendant la phase chronique subséquente). Les principaux facteurs cardiométaboliques dont les associations épidémiologiques et mécanistiques avec la COVID-19 ont été avérées comprennent l'adiposité anormale, la dysglycémie, la dyslipidémie et l'hypertension. L'hypertension suscite un intérêt particulier, car certaines composantes du système rénine-angiotensine (SRA), dont le rôle est crucial dans la physiopathologie de l'hypertension, sont également en cause dans la COVID-19. Plus précisément, l'enzyme de conversion de l'angiotensine 2 (ECA2), une protéine multifonctionnelle du SRA faisant partie de l'axe protecteur du SRA, est également le récepteur permettant au virus SRAS-CoV-2 d'entrer dans les cellules hôtes et de provoquer une infection virale. Les affections cardiovasculaires et cardiométaboliques concomitantes ne font pas que prédisposer les personnes qui en sont atteintes à la COVID-19, elles constituent également des complications de l'infection à SRAS-CoV-2. En outre, de plus en plus de données probantes indiquent que l'atteinte rénale aiguë est fréquente en cas de COVID-19, qu'elle survient tôt et fait l'objet d'une association temporelle avec l'insuffisance respiratoire, et qu'elle est associée à un pronostic sombre, notamment en présence de facteurs de risque cardiovasculaires. Nous discutons ici des maladies cardiovasculaires et rénales dans le contexte de la COVID-19, et présentons les progrès récents sur les mécanismes physiopathologiques en cause dans le lien entre les maladies cardiovasculaires et la COVID-19 en nous attardant sur le SRA et l'ECA2, ainsi que sur le système immunitaire et l'inflammation. Nous présentons de l'information à jour sur les liens entre l'hypertension, le diabète et la COVID-19, et soulignons les principales maladies cardiovasculaires associées à la COVID-19. Nous analysons également brièvement les complications cardiovasculaires émergentes associées à la COVID-19 de longue durée, notamment le syndrome de tachycardie orthostatique posturale (STOP).
- Published
- 2021
15. Selective inhibition of the C-domain of ACE (angiotensin-converting enzyme) combined with inhibition of NEP (neprilysin): a potential new therapy for hypertension
- Author
-
Augusto C. Montezano, Tomasz J. Guzik, Karla B Neves, Marko Poglitsch, Francisco J. Rios, Lauren B. Arendse, Rhian M. Touyz, Delyth Graham, Rheure Alves-Lopes, Dominik Skiba, Adam Harvey, and Edward D. Sturrock
- Subjects
0301 basic medicine ,Pyridines ,Thiazepines ,medicine.drug_class ,Antihypertensive Treatment ,Angiotensin-Converting Enzyme Inhibitors ,Blood Pressure ,Mice, Transgenic ,Vascular permeability ,030204 cardiovascular system & hematology ,Pharmacology ,Sacubitril ,neprilysin ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Lisinopril ,Renin ,Internal Medicine ,medicine ,Animals ,Antihypertensive drug ,Antihypertensive Agents ,omapatrilat ,business.industry ,Aminobutyrates ,Biphenyl Compounds ,Body Weight ,Original Articles ,Angiotensin II ,vasodilatation ,030104 developmental biology ,Blood pressure ,Liver ,Hypertension ,ACE inhibitor ,ComputingMethodologies_DOCUMENTANDTEXTPROCESSING ,Omapatrilat ,permeability ,business ,medicine.drug - Abstract
Supplemental Digital Content is available in the text., Combined inhibition of NEP (neutral endopeptidase) and ACE (angiotensin-converting enzyme), without unwanted effects, remains an attractive therapeutic strategy in cardiovascular medicine. Omapatrilat, a dual NEP inhibitor–ACE inhibitor, was a promising antihypertensive drug but failed in trials due to angioedema, an effect possibly caused by inhibition of both the N- and C-domains of ACE. Here, we aimed to determine whether lisinopril-tryptophan (lisW-S), a C-domain specific ACE inhibitor that preserves the N-domain catalytic activity, together with sacubitril (NEP inhibitor), differentially influences cardiovascular function and vascular permeability in hypertension compared with omapatrilat and lisinopril+sacubitril which inhibits both the ACE C- and N-domains. Ang II (angiotensin II)–dependent hypertensive mice (transgenic mice expressing active human renin in the liver [also known as LinA3]) received vehicle, sacubitril, lisW-S, lisinopril, lisinopril+sacubitril, or lisW-S+sacubitril for 4 weeks. Systolic blood pressure was increased in LinA3 mice, along with cardiac hypertrophy/dysfunction, impaired endothelium-dependent vasorelaxation, hypercontractile responses, vascular remodeling, and renal inflammation. LisW-S+sacubitril, lisinopril+sacubitril, and omapatrilat reduced systolic blood pressure and normalized cardiovascular remodeling and vascular hypercontractile responses in LinA3 mice. Although lisinopril+sacubitril and omapatrilat improved Ach-induced vasorelaxation, lisW-S+sacubitril had no effect. Endothelial permeability (Evans Blue assessment) was increased in omapatrilat but not in LisW-S+sacubitril–treated mice. In conclusion, lisW-S combined with sacubitril reduced systolic blood pressure and improved cardiac dysfunction in LinA3 mice, similar to omapatrilat but without effects on endothelium-dependent vasorelaxation. Moreover, increased vascular leakage (plasma extravasation) induced by omapatrilat was not evident in mice treated with lisW-S+sacubitril. Targeting ACE C-domain and NEP as a combination therapy may be as effective as omapatrilat in lowering systolic blood pressure, but without inducing vascular permeability and endothelial injury.
- Published
- 2021
16. NOX5: Molecular biology and pathophysiology
- Author
-
Aikaterini Anagnostopoulou, Augusto C. Montezano, Francisco J. Rios, Rhian M. Touyz, and Livia L Camargo
- Subjects
Gene isoform ,Physiology ,kidney disease ,Nox isoforms ,030204 cardiovascular system & hematology ,medicine.disease_cause ,Gene Expression Regulation, Enzymologic ,03 medical and health sciences ,0302 clinical medicine ,cardiovascular disease ,Physiology (medical) ,medicine ,cancer ,oxidative stress ,Animals ,Humans ,Lecture ,reactive oxygen species ,Nutrition and Dietetics ,NADPH oxidase ,biology ,Endoplasmic reticulum ,Lymphocyte differentiation ,General Medicine ,Hydrogen-Ion Concentration ,Proton Pumps ,Cell biology ,Isoenzymes ,NADPH Oxidase 5 ,Dual oxidase 1 ,biology.protein ,030217 neurology & neurosurgery ,Function (biology) ,Oxidative stress ,Intracellular ,Genome-Wide Association Study - Abstract
New findings What is the topic of this review? This review provides a comprehensive overview of Nox5 from basic biology to human disease and highlights unique features of this Nox isoform What advances does it highlight? Major advances in Nox5 biology relate to crystallization of the molecule and new insights into the pathophysiological role of Nox5. Recent discoveries have unravelled the crystal structure of Nox5, the first Nox isoform to be crystalized. This provides new opportunities to develop drugs or small molecules targeted to Nox5 in an isoform-specific manner, possibly for therapeutic use. Moreover genome wide association studies (GWAS) identified Nox5 as a new blood pressure-associated gene and studies in mice expressing human Nox5 in a cell-specific manner have provided new information about the (patho) physiological role of Nox5 in the cardiovascular system and kidneys. Nox5 seems to be important in the regulation of vascular contraction and kidney function. In cardiovascular disease and diabetic nephropathy, Nox5 activity is increased and this is associated with increased production of reactive oxygen species and oxidative stress implicated in tissue damage. Abstract Nicotinamide adenine dinucleotide phosphate (NADPH) oxidases (Nox), comprise seven family members (Nox1-Nox5 and dual oxidase 1 and 2) and are major producers of reactive oxygen species in mammalian cells. Reactive oxygen species are crucially involved in cell signalling and function. All Noxs share structural homology comprising six transmembrane domains with two haem-binding regions and an NADPH-binding region on the intracellular C-terminus, whereas their regulatory systems, mechanisms of activation and tissue distribution differ. This explains the diverse function of Noxs. Of the Noxs, NOX5 is unique in that rodents lack the gene, it is regulated by Ca2+ , it does not require NADPH oxidase subunits for its activation, and it is not glycosylated. NOX5 localizes in the perinuclear and endoplasmic reticulum regions of cells and traffics to the cell membrane upon activation. It is tightly regulated through numerous post-translational modifications and is activated by vasoactive agents, growth factors and pro-inflammatory cytokines. The exact pathophysiological significance of NOX5 remains unclear, but it seems to be important in the physiological regulation of sperm motility, vascular contraction and lymphocyte differentiation, and NOX5 hyperactivation has been implicated in cardiovascular disease, kidney injury and cancer. The field of NOX5 biology is still in its infancy, but with new insights into its biochemistry and cellular regulation, discovery of the NOX5 crystal structure and genome-wide association studies implicating NOX5 in disease, the time is now ripe to advance NOX5 research. This review provides a comprehensive overview of our current understanding of NOX5, from basic biology to human disease, and highlights the unique characteristics of this enigmatic Nox isoform.
- Published
- 2019
17. ACE2 and Proinflammatory Signaling by S1 Protein of SARS‐Cov‐2 in Human Endothelial Cells
- Author
-
Wendy Beattie, Augusto C. Montezano, Livia L Camargo, Rheure A Lopes, Stuart A. Nicklin, Karla B Neves, Colin Berry, Rhian M. Touyz, and Francisco J. Rios
- Subjects
Pharmacology ,Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) ,Genetics ,Biology ,Molecular Biology ,Biochemistry ,Virology ,Pharmacology ‐ COVID‐19 ,Biotechnology ,Proinflammatory cytokine - Abstract
Introduction COVID‐19 is primarily a respiratory disease associated with cardiovascular risk. SARS‐CoV‐2, the virus causing COVID‐19, uses ACE2, an important enzyme in the cardiovascular system that regulates the conversion of Ang II (deleterious/pro‐hypertensive) to Ang 1‐7 (protective/anti‐hypertensive), as a receptor for host cell entry and infection. Considering the relationship between the viral S1‐protein and the host's ACE2, it is unclear whether this interaction is merely a mechanism of infection or whether it also contributes to cardiovascular damage associated with COVID‐19. We hypothesisedthat SARS‐Cov‐2‐ACE2 interaction induces activation of vascular cell inflammatory responses that are influenced by ACE2 dependent and/or independent enzymatic Ang‐(1‐7) production. Methods Human microvascular endothelial cells (MEC) were used and stimulated with SARS‐CoV‐2 recombinant S1 protein (rS1p) (0.66 μg/mL) at 10/30 min (acute) and 5/24h (chronic). Activation of pro‐inflammatory signaling pathways (immunoblotting, real‐time PCR), microparticle (MP) generation (NanoSight), and cytokine production (ELISA) were assessed. In some experiments, cells were pre‐incubated with an ACE2 activator (DIZE – 190 nM) and inhibitor (MLN‐4760 – 440 pM). Results rS1P increased NFκB activation (Control ©=0.99±0.06 vs. 1.38±0.19 AU; p
- Published
- 2021
18. The magnesium global network (MaGNet) to promote research on magnesium in diseases focusing on covid-19
- Author
-
Federica I, Wolf, Jeanette A, Maier, Andrea, Rosanoff, Mario, Barbagallo, Shadi, Baniasadi, Sara, Castiglioni, Fu-Chou, Cheng, Sherrie Colaneri, Day, Rebecca B, Costello, Ligia J, Dominguez, Ronald J, Elin, Claudia, Gamboa-Gomez, Fernando, Guerrero-Romero, Ka, Kahe, Klaus, Kisters, Martin, Kolisek, Anton, Kraus, Stefano, Iotti, Andre, Mazur, Moises, Mercado-Atri, Lucia, Merolle, Oliver, Micke, Nana, Gletsu-Miller, Forrest, Nielsen, Jin, O-Uchi, Ornella, Piazza, Michael, Plesset, Guitti, Pourdowlat, Francisco J, Rios, Martha, Rodriguez-Moran, Giuliana, Scarpati, Michael, Shechter, Yiqing, Song, Lisa A, Spence, Rhian M, Touyz, Valentina, Trapani, Nicola, Veronese, Bodo, von Ehrlich, Juergen, Vormann, Taylor C, Wallace, Cmer Center For Magnesium Education Research, Gesellschaft Für Magnesium-Forschung E V Germany, Sdrm Society International Society For The Development Of Research On Magnesium, Wolf F.I., Maier J.A., Rosanoff A., Barbagallo M., Baniasadi S., Castiglioni S., Cheng F.-C., Day S.C., Costello R.B., Dominguez L.J., Elin R.J., Gamboa-Gomez C., Guerrero-Romero F., Kahe K., Kisters K., Kolisek M., Kraus A., Iotti S., Mazur A., Mercado-Atri M., Merolle L., Micke O., Gletsu-Miller N., Nielsen F., O-Uchi J., Piazza O., Plesset M., Pourdowlat G., Rios F.J., Rodriguez-Moran M., Scarpati G., Shechter M., Song Y., Spence L.A., Touyz R.M., Trapani V., Veronese N., Von Ehrlich B., Vormann J., Wallace T.C., and Wolf FI, Maier JA, Rosanoff A, Barbagallo M, Baniasadi S, Castiglioni S, Cheng FC, Day SC, Costello RB, Dominguez LJ, Elin RJ, Gamboa-Gomez C, Guerrero-Romero F, Kahe K, Kisters K, Kolisek M, Kraus A, Iotti S, Mazur A, Mercado-Atri M, Merolle L, Micke O, Gletsu-Miller N, Nielsen F, O-Uchi J, Piazza O, Plesset M, Pourdowlat G, Rios FJ, Rodriguez-Moran M, Scarpati G, Shechter M, Song Y, Spence LA, Touyz RM, Trapani V, Veronese N, von Ehrlich B, Vormann J, Wallace TC
- Subjects
Societies, Scientific ,Aging ,Supplementation ,Comorbidity ,Metabolic Diseases ,Settore MED/04 - PATOLOGIA GENERALE ,Neoplasms ,Hypomagnesaemia ,Cardiovascular Disease ,Humans ,Magnesium ,Obesity ,Disease severity ,Nutrition ,Inflammation ,Prevention ,Research ,COVID-19 ,Thrombosis ,Scientific ,Hypermagnesaemia ,Congresses as Topic ,Metabolic Disease ,Cardiovascular Diseases ,Immune System ,ICU ,Thrombosi ,Neoplasm ,Disease Susceptibility ,hypomagnesaemia, hypermagnesaemia, inflammation, thrombosis, prevention, disease severity, supplementation, nutrition, ICU ,Societies ,Magnesium Deficiency ,Human - Abstract
When the current SARS-CoV-2 pandemic began in early 2020, the global magnesium researcher community came together and noted the striking similarities between COVID-19 risk factors and conditions associated with magnesium deficit state in humans, reasoning that magnesium deficiency could worsen the course of COVID-19 [1-4]. This prompted establishment of a worldwide collaborative network with regular virtual meetings to brainstorm the associations between magnesium and COVID-19. We hypothesize that magnesium deficiency, a common but mostly unrecognized state in modern global societies, could be an important component of the susceptibility to SARS-CoV-2 infection. Consequently, restoring the magnesium deficit may be a putative therapeutic strategy to possibly ameliorate or prevent COVID-19.
- Published
- 2021
19. Oxidative stress: a unifying paradigm in hypertension
- Author
-
Augusto C. Montezano, Livia L Camargo, Karla B Neves, Rheure Alves-Lopes, Francisco J. Rios, and Rhian M. Touyz
- Subjects
Inflammation ,Vascular Remodeling ,medicine.disease_cause ,Article ,Immune system ,Mediator ,Sex Factors ,Vascular Stiffness ,Fibrosis ,medicine ,Humans ,Aldosterone ,chemistry.chemical_classification ,Reactive oxygen species ,Endothelin-1 ,business.industry ,Angiotensin II ,Inflammasome ,medicine.disease ,Cell biology ,Oxidative Stress ,chemistry ,Vasoconstriction ,Hypertension ,Intercellular Signaling Peptides and Proteins ,Endothelium, Vascular ,Signal transduction ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Reactive Oxygen Species ,Oxidation-Reduction ,Oxidative stress ,medicine.drug ,Signal Transduction - 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.
- Published
- 2020
20. Acute effects of electronic and tobacco cigarettes on vascular and respiratory function in healthy volunteers: a cross-over study
- Author
-
Francisco J. Rios, Richard G. Taylor, Karine Pinel, Rhian M. Touyz, Christian Delles, Katriona Brooksbank, and Danièle M I Kerr
- Subjects
medicine.medical_specialty ,Physiology ,Blood Pressure ,030204 cardiovascular system & hematology ,Cigarette Smoking ,03 medical and health sciences ,Hyperaemia ,0302 clinical medicine ,Cell-Derived Microparticles ,Heart Rate ,Internal medicine ,Heart rate ,Internal Medicine ,medicine ,Humans ,Respiratory function ,030212 general & internal medicine ,Cross-Over Studies ,business.industry ,Respiration ,Vaping ,medicine.disease ,Thrombosis ,Crossover study ,Blood pressure ,Cardiology ,Arterial stiffness ,Biomarker (medicine) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business - Abstract
Objectives: To assess the acute effects of nicotine-containing electronic cigarettes versus tobacco smoking on vascular and respiratory function and circulating microparticles, particularly platelet microparticles (PMPs, biomarker of haemostasis/thrombosis) and endothelial microparticles (EMPs, biomarker of endothelial function).\ud \ud Methods: Heart rate (HR), blood pressure, reactive hyperaemia index (RHI, microvascular reactivity), augmentation index (arterial stiffness) and respiratory function were assessed in 20 smokers immediately before and after electronic cigarettes use and tobacco smoking. The number of microparticles was determined by flow cytometry using counting beads as a reference. Labelling with Annexin-V was used to detect the total microparticle fraction. EMPs were characterized as CD31+CD42− and PMPs as CD31+CD42+.\ud \ud Results: HR increased after electronic cigarettes use and tobacco smoking (P < 0.001), whereas blood pressure remained unchanged (P > 0.05). RHI (P = 0.006), augmentation index (P = 0.010) but not augmentation index standardized to HR 75 bpm (P > 0.05) increased with electronic cigarettes use but not with tobacco smoking. Following tobacco smoking, there was a significant increase in total microparticles (P < 0.001), EMPs (P < 0.001) and PMPs (P < 0.001). In contrast, electronic cigarettes were only associated with an increase in PMPs (P < 0.001), with no significant changes in the total microparticle fraction or EMPs (all P > 0.05). Peak expiratory flow significantly decreased following electronic cigarettes use (P = 0.019).\ud \ud Conclusion: Our results demonstrate that acute exposure to tobacco smoking as well as electronic cigarettes influences vascular and respiratory function. Where tobacco smoking significantly increased microparticle formation, indicative of possible endothelial injury, electronic cigarettes use induced vasoreactivity and decreased peak expiratory flow. These findings suggest that both electronic cigarettes and tobacco smoking negatively impact vascular function.
- Published
- 2019
21. Interplay between Hormones, the Immune System, and Metabolic Disorders
- Author
-
Joilson O. Martins, Naima Moustaid-Moussa, and Francisco J. Rios
- Subjects
0301 basic medicine ,Article Subject ,business.industry ,Immunology ,Cell Biology ,Bioinformatics ,Hormones ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Editorial ,Metabolic Diseases ,Immune System ,lcsh:Pathology ,Medicine ,Animals ,Humans ,Hormone metabolism ,030212 general & internal medicine ,business ,Hormone ,lcsh:RB1-214 - Abstract
No abstract available.
- Published
- 2018
22. NADPH Oxidase 5 is a pro‐contractile Nox isoform and a point of cross‐talk for calcium and redox signaling‐implications in vascular function
- Author
-
Aikaterini Anagnostopoulou, Augusto C. Montezano, Ana Caroline P. Gandara, Delyth Graham, Roberto Palacios, Karla B Neves, Maria Dulak-Lis, Livia L Camargo, Rheure Alves-Lopes, Francisco J. Rios, Adam Harvey, Patrik Persson, Pedro Lagerblad de Oliveira, Chris R. J. Kennedy, Rhian M. Touyz, and Chet E. Holterman
- Subjects
0301 basic medicine ,Gene isoform ,Cell signaling ,Contraction (grammar) ,Heart Diseases ,Myocytes, Smooth Muscle ,chemistry.chemical_element ,Blood Pressure ,Mice, Transgenic ,Calcium ,Endoplasmic Reticulum ,Muscle, Smooth, Vascular ,03 medical and health sciences ,Calmodulin ,Medicine ,cell signaling ,Animals ,Humans ,Calcium Signaling ,NOx ,Cells, Cultured ,Original Research ,chemistry.chemical_classification ,Reactive oxygen species ,business.industry ,contraction ,vascular biology ,Cell biology ,Vasodilation ,Disease Models, Animal ,030104 developmental biology ,chemistry ,NADPH Oxidase 5 ,Vasoconstriction ,Rhodnius ,NADPH oxidase 5 ,Hypertension ,Insect Proteins ,Cardiology and Cardiovascular Medicine ,Vascular function ,business ,Reactive Oxygen Species ,Oxidation-Reduction ,Cell Signalling/Signal Transduction - Abstract
Background NADPH Oxidase 5 (Nox5) is a calcium‐sensitive superoxide‐generating Nox. It is present in lower forms and higher mammals, but not in rodents. Nox5 is expressed in vascular cells, but the functional significance remains elusive. Given that contraction is controlled by calcium and reactive oxygen species, both associated with Nox5, we questioned the role of Nox5 in pro‐contractile signaling and vascular function. Methods and Results Transgenic mice expressing human Nox5 in a vascular smooth muscle cell–specific manner (Nox5 mice) and Rhodnius prolixus , an arthropod model that expresses Nox5 endogenoulsy, were studied. Reactive oxygen species generation was increased systemically and in the vasculature and heart in Nox5 mice. In Nox5‐expressing mice, agonist‐induced vasoconstriction was exaggerated and endothelium‐dependent vasorelaxation was impaired. Vascular structural and mechanical properties were not influenced by Nox5. Vascular contractile responses in Nox5 mice were normalized by N ‐acetylcysteine and inhibitors of calcium channels, calmodulin, and endoplasmic reticulum ryanodine receptors, but not by GKT 137831 (Nox1/4 inhibitor). At the cellular level, vascular changes in Nox5 mice were associated with increased vascular smooth muscle cell [Ca 2+ ] i , increased reactive oxygen species and nitrotyrosine levels, and hyperphosphorylation of pro‐contractile signaling molecules MLC 20 (myosin light chain 20) and MYPT 1 (myosin phosphatase target subunit 1). Blood pressure was similar in wild‐type and Nox5 mice. Nox5 did not amplify angiotensin II effects. In R. prolixus , gastrointestinal smooth muscle contraction was blunted by Nox5 silencing, but not by VAS 2870 (Nox1/2/4 inhibitor). Conclusions Nox5 is a pro‐contractile Nox isoform important in redox‐sensitive contraction. This involves calcium‐calmodulin and endoplasmic reticulum–regulated mechanisms. Our findings define a novel function for vascular Nox5, linking calcium and reactive oxygen species to the pro‐contractile molecular machinery in vascular smooth muscle cells.
- Published
- 2018
23. Boosting Adaptive Immunity: A New Role for PAFR Antagonists
- Author
-
Marianna M. Koga, Sonia Jancar, Bruna Bizzarro, Francisco J. Rios, and Anderson Sá-Nunes
- Subjects
0301 basic medicine ,CD4-Positive T-Lymphocytes ,Ovalbumin ,medicine.medical_treatment ,T cell ,Freund's Adjuvant ,Spleen ,chemical and pharmacologic phenomena ,Platelet Membrane Glycoproteins ,Biology ,Adaptive Immunity ,Article ,Receptors, G-Protein-Coupled ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Immune system ,In vivo ,medicine ,Splenocyte ,Animals ,IMUNOLOGIA ,Cells, Cultured ,Multidisciplinary ,FOXP3 ,Azepines ,Dendritic Cells ,Triazoles ,Acquired immune system ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Immunoglobulin G ,Immunology ,Immunization ,Adjuvant ,Platelet Aggregation Inhibitors - Abstract
We have previously shown that the Platelet-Activating Factor Receptor (PAFR) engagement in murine macrophages and dendritic cells (DCs) promotes a tolerogenic phenotype reversed by PAFR-antagonists treatment in vitro. Here, we investigated whether a PAFR antagonist would modulate the immune response in vivo. Mice were subcutaneously injected with OVA or OVA with PAFR-antagonist WEB2170 on days 0 and 7. On day 14, OVA–specific IgG2a and IgG1 were measured in the serum. The presence of WEB2170 during immunization significantly increased IgG2a without affecting IgG1 levels. When WEB2170 was added to OVA in complete Freund’s adjuvant, enhanced IgG2a but not IgG1 production was also observed, and CD4+ FoxP3+ T cell frequency in the spleen was reduced compared to mice immunized without the antagonist. Similar results were observed in PAFR-deficient mice, along with increased Tbet mRNA expression in the spleen. Additionally, bone marrow-derived DCs loaded with OVA were transferred into naïve mice and their splenocytes were co-cultured with fresh OVA-loaded DCs. CD4+ T cell proliferation was higher in the group transferred with DCs treated with the PAFR-antagonist. We propose that the activation of PAFR by ligands present in the site of immunization is able to fine-tune the adaptive immune response.
- Published
- 2016
- Full Text
- View/download PDF
24. Reply to letter by McIntyre et al
- Author
-
Rhian M. Touyz, Augusto C. Montezano, Francisco J. Rios, and Carmine Savoia
- Subjects
Vascular Endothelial Growth Factor A ,receptorantineoplastic ,business.industry ,bevacizumabendothelial ,nitric oxide ,synthasenitric ,oxidevasculotropinvasculotropin ,agentvasculotropin A ,Disease Management ,Antineoplastic Agents ,Neoplasms ,Hypertension ,Medicine ,Humans ,Cardiology and Cardiovascular Medicine ,business ,Humanities - Published
- 2014
25. Hypertension Due to Antiangiogenic Cancer Therapy With Vascular Endothelial Growth Factor Inhibitors: Understanding and Managing a New Syndrome
- Author
-
Augusto C. Montezano, Francisco J. Rios, Heather Y Small, Rhian M. Touyz, and Carmine Savoia
- Subjects
medicine.medical_specialty ,business.industry ,Dihydropyridine ,Cancer ,Prostacyclin ,Disease ,medicine.disease ,Bioinformatics ,Nitric oxide ,Vascular endothelial growth factor ,chemistry.chemical_compound ,Endocrinology ,Blood pressure ,chemistry ,Internal medicine ,medicine ,Endothelial dysfunction ,Cardiology and Cardiovascular Medicine ,business ,medicine.drug - Abstract
Novel antiangiogenic cancer therapies, particularly agents that block vascular endothelial growth factor (VEGF) signalling, have improved outcomes in patients with cancers and are now used as first-line therapies for some tumours. However, with VEGF inhibitors (VEGFIs) are new complications, particularly hypertension. VEGFI-induced hypertension is a dose-dependent phenomenon due to on-target effects rather than off-target effects. Increased blood pressure occurs in almost 100% of patients who take VEGFIs, with a subset who develop severe hypertension. Molecular mechanisms underlying VEGFI-induced hypertension are unclear, but endothelial dysfunction and increased vascular resistance, due to impaired nitric oxide signalling, reduced prostacyclin production, endothelin-1 (ET-1) upregulation, oxidative stress, and rarefaction have been implicated. Treatment of hypertension should be aimed at reducing the risk of short-term morbidity associated with hypertension while maintaining effective dosing of antiangiogenic therapy for optimal cancer treatment. Although specific guidelines are not yet available for the management of VEGFI-induced hypertension, angiotensin-converting enzyme inhibitors and dihydropyridine calcium channel blockers are commonly used. Severe hypertension might require reduction of VEGFI dosing, or in some cases, interruption of treatment. As more potent VEGFIs are developed and as more cancer patients are treated with VEGFIs, the burden of hypertension toxicity will increase. This will be further compounded as the use of antiangiogenic drugs broadens to include older patients and those with pre-existing cardiovascular disease. Here we focus on VEGF as a target for antiangiogenesis and how this affects increased blood pressure. Putative mechanisms underlying VEGFI-induced hypertension are highlighted and therapeutic strategies to manage such hypertension are discussed.
- Published
- 2014
26. Clearance of Apoptotic Cells by Macrophages Induces Regulatory Phenotype and Involves Stimulation of CD36 and Platelet-Activating Factor Receptor
- Author
-
Sonia Jancar, Mateus Pecenin, Francisco J. Rios, and Matheus Ferracini
- Subjects
CD36 Antigens ,Male ,Article Subject ,CD36 ,Phagocytosis ,Immunology ,Apoptosis ,Platelet Membrane Glycoproteins ,Receptors, G-Protein-Coupled ,Mice ,lcsh:Pathology ,Macrophage ,Animals ,Efferocytosis ,Lipid raft ,IMUNOLOGIA ,biology ,Interleukin-12 Subunit p40 ,Macrophages ,Cell Biology ,Cell biology ,Interleukin-10 ,Mice, Inbred C57BL ,Interleukin 10 ,Phenotype ,biology.protein ,Platelet-activating factor receptor ,Research Article ,lcsh:RB1-214 - Abstract
Phagocytosis of apoptotic cells (efferocytosis) induces macrophage differentiation towards a regulatory phenotype (IL-10high/IL-12p40low). CD36 is involved in the recognition of apoptotic cells (AC), and we have shown that the platelet-activating factor receptor (PAFR) is also involved. Here, we investigated the contribution of PAFR and CD36 to efferocytosis and to the establishment of a regulatory macrophage phenotype. Mice bone marrow-derived macrophages were cocultured with apoptotic thymocytes, and the phagocytic index was determined. Blockage of PAFR with antagonists or CD36 with specific antibodies inhibited the phagocytosis of AC (~70–80%). Using immunoprecipitation and confocal microscopy, we showed that efferocytosis increased the CD36 and PAFR colocalisation in the macrophage plasma membrane; PAFR and CD36 coimmunoprecipitated with flotillin-1, a constitutive lipid raft protein, and disruption of these membrane microdomains by methyl-β-cyclodextrin reduced AC phagocytosis. Efferocytosis induced a pattern of cytokine production, IL-10high/IL-12p40low, that is, characteristic of a regulatory phenotype. LPS potentiated the efferocytosis-induced production of IL-10, and this was prevented by blocking PAFR or CD36. It can be concluded that phagocytosis of apoptotic cells engages CD36 and PAFR, possibly in lipid rafts, and this is required for optimal efferocytosis and the establishment of the macrophage regulatory phenotype.
- Published
- 2013
27. Oxidative stress and modification of renal vascular permeability are associated with acute kidney injury during P. berghei ANKA infection
- Author
-
Meire Ioshie Hyane, Reinaldo Correia Silva, Renato Barboza, Claudio Romero Farias Marinho, Rosa Maria Elias, Marlene Antônia dos Reis, Tarcio Teodoro Braga, Niels Olsen Saraiva Câmara, Antônio Martins Figueiredo-Neto, Alvaro Pacheco-Silva, Ângela Castoldi, Matheus Correa-Costa, Giselle Martins Gonçalves, Claudiene Rodrigues Barreto, Francisco J. Rios, C. Y. Hayashida, Maria Regina D'Império-Lima, Marcos Antonio Cenedeze, and Alexandre C. Keller
- Subjects
Erythrocytes ,Mouse ,Plasmodium berghei ,PARASITOLOGIA ,Apoptosis ,Vascular permeability ,Kidney ,medicine.disease_cause ,Mice ,Molecular Cell Biology ,Immune Response ,Cellular Stress Responses ,Mice, Inbred BALB C ,Multidisciplinary ,Cell Death ,Acute kidney injury ,Animal Models ,Acute Kidney Injury ,Cell Hypoxia ,Infectious Diseases ,medicine.anatomical_structure ,Nephrology ,Medicine ,Research Article ,Science ,Immunology ,Renal function ,Heme ,Biology ,Proinflammatory cytokine ,Capillary Permeability ,Model Organisms ,parasitic diseases ,Cell Adhesion ,Parasitic Diseases ,medicine ,Animals ,Inflammation ,Immunity ,Endothelial Cells ,Kidney metabolism ,medicine.disease ,biology.organism_classification ,Malaria ,Oxidative Stress ,Acute Renal Failure ,Reactive Oxygen Species ,Oxidative stress - Abstract
Malaria associated-acute kidney injury (AKI) is associated with 45% of mortality in adult patients hospitalized with severe form of the disease. However, the causes that lead to a framework of malaria-associated AKI are still poorly characterized. Some clinical studies speculate that oxidative stress products, a characteristic of Plasmodium infection, as well as proinflammatory response induced by the parasite are involved in its pathophysiology. Therefore, we aimed to investigate the development of malaria-associated AKI during infection by P. berghei ANKA, with special attention to the role played by the inflammatory response and the involvement of oxidative stress. For that, we took advantage of an experimental model of severe malaria that showed significant changes in the renal pathophysiology to investigate the role of malaria infection in the renal microvascular permeability and tissue injury. Therefore, BALB/c mice were infected with P. berghei ANKA. To assess renal function, creatinine, blood urea nitrogen, and ratio of proteinuria and creatininuria were evaluated. The products of oxidative stress, as well as cytokine profile were quantified in plasma and renal tissue. The change of renal microvascular permeability, tissue hypoxia and cellular apoptosis were also evaluated. Parasite infection resulted in renal dysfunction. Furthermore, we observed increased expression of adhesion molecule, proinflammatory cytokines and products of oxidative stress, associated with a decrease mRNA expression of HO-1 in kidney tissue of infected mice. The measurement of lipoprotein oxidizability also showed a significant increase in plasma of infected animals. Together, our findings support the idea that products of oxidative stress, as well as the immune response against the parasite are crucial to changes in kidney architecture and microvascular endothelial permeability of BALB/c mice infected with P. berghei ANKA.
- Published
- 2012
28. Expression of PAFR as Part of a Prosurvival Response to Chemotherapy: A Novel Target for Combination Therapy in Melanoma
- Author
-
Camila Maria Longo Machado, Renata F. Saito, Sonia Jancar, Ana Cláudia Onuchic, Roger Chammas, and Francisco J. Rios
- Subjects
Programmed cell death ,Article Subject ,Combination therapy ,Cell Survival ,Immunology ,Mice, Nude ,Antineoplastic Agents ,Platelet Membrane Glycoproteins ,Biology ,Pharmacology ,Receptors, G-Protein-Coupled ,Mice ,Cell Line, Tumor ,Antineoplastic Combined Chemotherapy Protocols ,medicine ,lcsh:Pathology ,Animals ,Humans ,Receptor ,Melanoma ,Cisplatin ,Macrophages ,Azepines ,Cell Biology ,Triazoles ,medicine.disease ,In vitro ,Gene Expression Regulation, Neoplastic ,Cell culture ,Female ,Signal transduction ,Neoplasm Transplantation ,Signal Transduction ,Research Article ,medicine.drug ,lcsh:RB1-214 - Abstract
Melanoma cells express the platelet-activating factor receptor (PAFR) and, thus, respond to PAF, a bioactive lipid produced by both tumour cells and those in the tumour microenvironment such as macrophages. Here, we show that treatment of a human melanoma SKmel37 cell line with cisplatin led to increased expression of PAFR and its accumulation. In the presence of exogenous PAF, melanoma cells were significantly more resistant to cisplatin-induced cell death. Inhibition of PAFR-dependent signalling pathways by a PAFR antagonist (WEB2086) showed chemosensitisation of melanoma cellsin vitro. Nude mice were inoculated with SKmel37 cells and treated with cisplatin and WEB2086. Animals treated with both agents showed significantly decreased tumour growth compared to the control group and groups treated with only one agent. PAFR accumulation and signalling are part of a prosurvival program of melanoma cells, therefore constituting a promising target for combination therapy for melanomas.
- Published
- 2012
29. SARS-CoV-2 spike protein induces endothelial inflammation via ACE2 independently of viral replication
- Author
-
Augusto C. Montezano, Livia L. Camargo, Sheon Mary, Karla B Neves, Francisco J Rios, Ross Stein, Rheure A. Lopes, Wendy Beattie, Jacqueline Thomson, Vanessa Herder, Agnieszka M. Szemiel, Steven McFarlane, Massimo Palmarini, and Rhian M. Touyz
- Subjects
Medicine ,Science - Abstract
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.
- Published
- 2023
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.