Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Acta Pharmacol Sin ; 43(9): 2173-2190, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35046517

RESUMO

Colchicine is an ancient herbal drug derived from Colchicum autumnale. It was first used to treat familial Mediterranean fever and gout. Based on its unique efficacy as an anti-inflammatory agent, colchicine has been used in the therapy of cardiovascular diseases including coronary artery disease, atherosclerosis, recurrent pericarditis, vascular restenosis, heart failure, and myocardial infarction. More recently, colchicine has also shown therapeutic efficacy in alleviating cardiovascular complications of COVID-19. COLCOT and LoDoCo2 are two milestone clinical trials that confirm the curative effect of long-term administration of colchicine in reducing the incidence of cardiovascular events in patients with coronary artery disease. There is growing interest in studying the anti-inflammatory mechanisms of colchicine. The anti-inflammatory action of colchicine is mediated mainly through inhibiting the assembly of microtubules. At the cellular level, colchicine inhibits the following: (1) endothelial cell dysfunction and inflammation; (2) smooth muscle cell proliferation and migration; (3) macrophage chemotaxis, migration, and adhesion; (4) platelet activation. At the molecular level, colchicine reduces proinflammatory cytokine release and inhibits NF-κB signaling and NLRP3 inflammasome activation. In this review, we summarize the current clinical trials with proven curative effect of colchicine in treating cardiovascular diseases. We also systematically discuss the mechanisms of colchicine action in cardiovascular therapeutics. Altogether, colchicine, a bioactive constituent from an ancient medicinal herb, exerts unique anti-inflammatory effects and prominent cardiovascular actions, and will charter a new page in cardiovascular medicine.


Assuntos
Tratamento Farmacológico da COVID-19 , Fármacos Cardiovasculares , Doença da Artéria Coronariana , Infarto do Miocárdio , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Fármacos Cardiovasculares/farmacologia , Fármacos Cardiovasculares/uso terapêutico , Colchicina/farmacologia , Colchicina/uso terapêutico , Doença da Artéria Coronariana/tratamento farmacológico , Humanos , Infarto do Miocárdio/tratamento farmacológico
2.
Clin Sci (Lond) ; 131(12): 1345-1360, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-28487469

RESUMO

Phosphoinositide 3-kinase [PI3K (p110α)] is able to negatively regulate the diabetes-induced increase in NADPH oxidase in the heart. Patients affected by diabetes exhibit significant cardiovascular morbidity and mortality, at least in part due to a cardiomyopathy characterized by oxidative stress and left ventricular (LV) dysfunction. Thus, PI3K (p110α) may represent a novel approach to protect the heart from diabetes-induced cardiac oxidative stress and dysfunction. In the present study, we investigated the therapeutic potential of a delayed intervention with cardiac-targeted PI3K gene therapy, administered to mice with established diabetes-induced LV diastolic dysfunction. Diabetes was induced in 6-week-old male mice by streptozotocin (STZ). After 8 weeks of untreated diabetes, LV diastolic dysfunction was confirmed by a reduction in echocardiography-derived transmitral E/A ratio. Diabetic and non-diabetic mice were randomly allocated to receive either recombinant adeno-associated viral vector-6 carrying a constitutively-active PI3K construct (recombinant adeno-associated-virus 6-constitutively active PI3K (p110α) (caPI3K) (rAAV6-caPI3K), single i.v. injection, 2 × 1011 vector genomes) or null vector, and were followed for a further 6 or 8 weeks. At study endpoint, diabetes-induced LV dysfunction was significantly attenuated by a single administration of rAAV6-caPI3K, administered 8 weeks after the induction of diabetes. Diabetes-induced impairments in each of LV NADPH oxidase, endoplasmic reticulum (ER) stress, apoptosis, cardiac fibrosis and cardiomyocyte hypertrophy, in addition to LV systolic dysfunction, were attenuated by delayed intervention with rAAV6-caPI3K. Hence, our demonstration that cardiac-targeted PI3K (p110α) gene therapy limits diabetes-induced up-regulation of NADPH oxidase and cardiac remodelling suggests new insights into promising approaches for the treatment of diabetic cardiomyopathy, at a clinically relevant time point (after diastolic dysfunction is manifested).


Assuntos
Cardiomiopatias Diabéticas/prevenção & controle , Terapia Genética/métodos , Miocárdio/enzimologia , NADPH Oxidases/metabolismo , Fosfatidilinositol 3-Quinase/biossíntese , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda , Animais , Dependovirus/genética , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/enzimologia , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/fisiopatologia , Diástole , Vetores Genéticos , Masculino , Camundongos , Miocárdio/patologia , Fosfatidilinositol 3-Quinase/genética , Transdução de Sinais , Fatores de Tempo , Transdução Genética , Disfunção Ventricular Esquerda/enzimologia , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/fisiopatologia , Remodelação Ventricular
3.
Mucosal Immunol ; 2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38614323

RESUMO

Excessive inflammatory responses are the main characteristic of ulcerative colitis (UC). Activation of formyl peptide receptor 1 (FPR1) has been found to promote the proliferation and migration of epithelial cells, but its role and therapeutic potential in UC remain unclear. This study observed an increased expression of FPR1 in a mouse model of colitis. Interestingly, FPR1 deficiency exacerbated UC and increased the secretion of the proinflammatory mediator from immune cells (e.g. macrophages), S100a8, a member of the damage-associated molecular patterns. Notably, the administration of the FPR agonist Cmpd43 ameliorated colon injury in a preclinical mice model of UC, likely via inhibiting phosphorylation of cyclic adenosine monophosphate-response element-binding protein and expression of CCAAT/enhancer-binding protein ß, which in turn suppressed the secretion of S100a8. In conclusion, these findings discovered a novel role of FPR1 in the development of colitis and will facilitate the development of FPR1-based pharmacotherapy to treat UC.

4.
Am J Respir Cell Mol Biol ; 48(3): 346-53, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23239497

RESUMO

In asthma, basic fibroblast growth factor (FGF-2) plays an important (patho)physiological role. This study examines the effects of FGF-2 on the transforming growth factor-ß (TGF-ß)-stimulated differentiation of airway smooth muscle (ASM) cells in vitro. The differentiation of human ASM cells after incubation with TGF-ß (100 pM) and/or FGF-2 (300 pM) for 48 hours was assessed by increases in contractile protein expression, actin-cytoskeleton reorganization, enhancements in cell stiffness, and collagen remodeling. FGF-2 inhibited TGF-ß-stimulated increases in transgelin (SM22) and calponin gene expression (n = 15, P < 0.01) in an extracellular signal-regulated kinase 1/2 (ERK1/2) signal transduction-dependent manner. The abundance of ordered α-smooth muscle actin (α-SMA) filaments formed in the presence of TGF-ß were also reduced by FGF-2, as was the ratio of F-actin to G-actin (n = 8, P < 0.01). Furthermore, FGF-2 attenuated TGF-ß-stimulated increases in ASM cell stiffness and the ASM-mediated contraction of lattices, composed of collagen fibrils (n = 5, P < 0.01). However, the TGF-ß-stimulated production of IL-6 was not influenced by FGF-2 (n = 4, P > 0.05), suggesting that FGF-2 antagonism is selective for the regulation of ASM cell contractile protein expression, organization, and function. Another mitogen, thrombin (0.3 U ml(-1)), exerted no effect on TGF-ß-regulated contractile protein expression (n = 8, P > 0.05), α-SMA organization, or the ratio of F-actin to G-actin (n = 4, P > 0.05), suggesting that the inhibitory effect of FGF-2 is dissociated from its mitogenic actions. The addition of FGF-2, 24 hours after TGF-ß treatment, still reduced contractile protein expression, even when the TGF-ß-receptor kinase inhibitor, SB431542 (10 µM), was added 1 hour before FGF-2. We conclude that the ASM cell differentiation promoted by TGF-ß is antagonized by FGF-2. A better understanding of the mechanism of action for FGF-2 is necessary to develop a strategy for therapeutic exploitation in the treatment of asthma.


Assuntos
Diferenciação Celular/genética , Fator 2 de Crescimento de Fibroblastos/metabolismo , Músculo Liso/citologia , Miócitos de Músculo Liso/citologia , Sistema Respiratório/citologia , Sistema Respiratório/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Actinas/genética , Actinas/metabolismo , Asma/genética , Asma/metabolismo , Asma/patologia , Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao Cálcio/metabolismo , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Proteínas Contráteis/genética , Proteínas Contráteis/metabolismo , Fator 2 de Crescimento de Fibroblastos/genética , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Proteínas dos Microfilamentos/genética , Proteínas dos Microfilamentos/metabolismo , Proteínas Musculares/genética , Proteínas Musculares/metabolismo , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Fator de Resposta Sérica/genética , Fator de Resposta Sérica/metabolismo , Transdução de Sinais/genética , Fator de Crescimento Transformador beta/genética , Proteínas Elk-1 do Domínio ets/genética , Proteínas Elk-1 do Domínio ets/metabolismo , Calponinas
5.
Am J Respir Cell Mol Biol ; 49(5): 751-8, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23721211

RESUMO

Plasminogen has a role in airway inflammation. Airway smooth muscle (ASM) cells cleave plasminogen into plasmin, a protease with proinflammatory activity. In this study, the effect of plasminogen on cytokine production by human ASM cells was investigated in vitro. Levels of IL-6 and IL-8 in the medium of ASM cells were increased by incubation with plasminogen (5-50 µg/ml) for 24 hours (P < 0.05; n = 6-9), corresponding to changes in the levels of cytokine mRNA at 4 hours. The effects of plasminogen were attenuated by α2-antiplasmin (1 µg/ml), a plasmin inhibitor (P < 0.05; n = 6-12). Exogenous plasmin (5-15 mU/ml) also stimulated cytokine production (P < 0.05; n = 6-8) in a manner sensitive to serine-protease inhibition by aprotinin (10 KIU/ml). Plasminogen-stimulated cytokine production was increased in cells pretreated with basic fibroblast growth factor (300 pM) in a manner associated with increases in urokinase plasminogen activator expression and plasmin formation. The knockdown of annexin A2, a component of the putative plasminogen receptor comprised of annexin A2 and S100A10, attenuated plasminogen conversion into plasmin and plasmin-stimulated cytokine production by ASM cells. Moreover, a role for annexin A2 in airway inflammation was demonstrated in annexin A2-/- mice in which antigen-induced increases in inflammatory cell number and IL-6 levels in the bronchoalveolar lavage fluid were reduced (P < 0.01; n = 10-14). In conclusion, plasminogen stimulates ASM cytokine production in a manner regulated by annexin A2. Our study shows for the first time that targeting annexin A2-mediated signaling may provide a novel therapeutic approach to the treatment of airway inflammation in diseases such as chronic asthma.


Assuntos
Anexina A2/metabolismo , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Músculo Liso/metabolismo , Miócitos de Músculo Liso/metabolismo , Plasminogênio/metabolismo , Sistema Respiratório/metabolismo , Animais , Anexina A2/deficiência , Anexina A2/genética , Líquido da Lavagem Broncoalveolar/imunologia , Células Cultivadas , Citocinas/genética , Modelos Animais de Doenças , Fibrinolisina/metabolismo , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Músculo Liso/imunologia , Miócitos de Músculo Liso/imunologia , Fosfatidilinositol 3-Quinase/metabolismo , Pneumonia/imunologia , Pneumonia/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Mensageiro/metabolismo , Sistema Respiratório/imunologia , Transdução de Sinais , Fatores de Tempo , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , alfa 2-Antiplasmina/metabolismo
6.
Am J Physiol Heart Circ Physiol ; 305(3): H365-77, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23729209

RESUMO

Nitroxyl (HNO) is a redox congener of NO. We now directly compare the antihypertrophic efficacy of HNO and NO donors in neonatal rat cardiomyocytes and compare their contributing mechanisms of actions in this setting. Isopropylamine-NONOate (IPA-NO) elicited concentration-dependent inhibition of endothelin-1 (ET1)-induced increases in cardiomyocyte size, with similar suppression of hypertrophic genes. Antihypertrophic IPA-NO actions were significantly attenuated by l-cysteine (HNO scavenger), Rp-8-pCTP-cGMPS (cGMP-dependent protein kinase inhibitor), and 1-H-(1,2,4)-oxodiazolo-quinxaline-1-one [ODQ; to target soluble guanylyl cyclase (sGC)] but were unaffected by carboxy-PTIO (NO scavenger) or CGRP8-37 (calcitonin gene-related peptide antagonist). Furthermore, IPA-NO significantly increased cardiomyocyte cGMP 3.5-fold (an l-cysteine-sensitive effect) and stimulated sGC activity threefold, without detectable NO release. IPA-NO also suppressed ET1-induced cardiomyocyte superoxide generation. The pure NO donor diethylamine-NONOate (DEA-NO) reproduced these IPA-NO actions but was sensitive to carboxy-PTIO rather than l-cysteine. Although IPA-NO stimulation of purified sGC was preserved under pyrogallol oxidant stress (in direct contrast to DEA-NO), cardiomyocyte sGC activity after either donor was attenuated by this stress. Excitingly IPA-NO also exhibited acute antihypertrophic actions in response to pressure overload in the intact heart. Together these data strongly suggest that IPA-NO protection against cardiomyocyte hypertrophy is independent of both NO and CGRP but rather utilizes novel HNO activation of cGMP signaling. Thus HNO acutely limits hypertrophy independently of NO, even under conditions of elevated superoxide. Development of longer-acting HNO donors may thus represent an attractive new strategy for the treatment of cardiac hypertrophy, as stand-alone and/or add-on therapy to standard care.


Assuntos
Cardiomegalia/tratamento farmacológico , Fármacos Cardiovasculares/uso terapêutico , GMP Cíclico/metabolismo , Hidrazinas/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Óxidos de Nitrogênio/metabolismo , Sistemas do Segundo Mensageiro/efeitos dos fármacos , Animais , Animais Recém-Nascidos , Antioxidantes/farmacologia , Cardiomegalia/genética , Cardiomegalia/metabolismo , Cardiomegalia/patologia , Células Cultivadas , Proteína Quinase Dependente de GMP Cíclico Tipo I/antagonistas & inibidores , Proteína Quinase Dependente de GMP Cíclico Tipo I/metabolismo , Relação Dose-Resposta a Droga , Endotelina-1/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação da Expressão Gênica , Guanilato Ciclase/antagonistas & inibidores , Guanilato Ciclase/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Doadores de Óxido Nítrico/farmacologia , Pirogalol/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/metabolismo , Guanilil Ciclase Solúvel , Fatores de Tempo
7.
Am J Physiol Heart Circ Physiol ; 304(5): H729-39, 2013 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-23262135

RESUMO

Rats selectively bred for low (LCR) or high (HCR) intrinsic running capacity simultaneously present with contrasting risk factors for cardiovascular and metabolic disease. However, the impact of these phenotypes on left ventricular (LV) morphology and microvascular function, and their progression with aging, remains unresolved. We tested the hypothesis that the LCR phenotype induces progressive age-dependent LV remodeling and impairments in microvascular function, glucose utilization, and ß-adrenergic responsiveness, compared with HCR. Hearts and vessels isolated from female LCR (n = 22) or HCR (n = 26) were studied at 12 and 35 wk. Nonselected N:NIH founder rats (11 wk) were also investigated (n = 12). LCR had impaired glucose tolerance and elevated plasma insulin (but not glucose) and body-mass at 12 wk compared with HCR, with early LV remodeling. By 35 wk, LV prohypertrophic and glucose transporter GLUT4 gene expression were up- and downregulated, respectively. No differences in LV ß-adrenoceptor expression or cAMP content between phenotypes were observed. Macrovascular endothelial function was predominantly nitric oxide (NO)-mediated in both phenotypes and remained intact in LCR for both age-groups. In contrast, mesenteric arteries microvascular endothelial function, which was impaired in LCR rats regardless of age. At 35 wk, endothelial-derived hyperpolarizing factor-mediated relaxation was impaired whereas the NO contribution to relaxation is intact. Furthermore, there was reduced ß2-adrenoceptor responsiveness in both aorta and mesenteric LCR arteries. In conclusion, diminished intrinsic exercise capacity impairs systemic glucose tolerance and is accompanied by progressive development of LV remodeling. Impaired microvascular perfusion is a likely contributing factor to the cardiac phenotype.


Assuntos
Envelhecimento/fisiologia , Circulação Coronária/fisiologia , Tolerância ao Exercício/fisiologia , Coração/fisiologia , Remodelação Ventricular/fisiologia , Envelhecimento/genética , Animais , Fatores Biológicos/metabolismo , Tolerância ao Exercício/genética , Feminino , Fibrose/fisiopatologia , Transportador de Glucose Tipo 4/genética , Transportador de Glucose Tipo 4/metabolismo , Resistência à Insulina/genética , Resistência à Insulina/fisiologia , Síndrome Metabólica/genética , Síndrome Metabólica/fisiopatologia , Microcirculação/fisiologia , Miócitos Cardíacos/fisiologia , Óxido Nítrico/metabolismo , Fenótipo , Ratos , Ratos Endogâmicos , Receptores Adrenérgicos beta/metabolismo , Transdução de Sinais/fisiologia , Resistência Vascular/fisiologia , Vasodilatação/fisiologia , Miosinas Ventriculares/genética , Miosinas Ventriculares/metabolismo
8.
Biomed Pharmacother ; 165: 115238, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37536036

RESUMO

Stimulation of the angiotensin II type 2 receptor (AT2R) evokes protective effects in various cardiovascular diseases. Thus, this study aimed to investigate the effects of AT2R stimulation, with or without AT1R blockade, in a model of hypertension with concomitant type 1 diabetes mellitus (T1DM). Spontaneously hypertensive rats (SHRs) were given either citrate or a single dose of streptozotocin (STZ; 55 mg/kg, i.p.) to induce diabetes. After 4 weeks of diabetes, animals were administered either a vehicle (saline), AT2R agonist, ß-Pro7Ang III (0.1 mg/kg/day via osmotic mini-pump), AT1R blocker, candesartan (2 mg/kg/day via drinking water), or a combination of both for a further 8 weeks. ß-Pro7Ang III treatment had no effect on blood pressure, but attenuated the significant increase in cardiac interstitial collagen and protein expression of fibrotic and inflammatory markers, and superoxide levels that was evident in diabetic SHRs. These effects were not observed with candesartan, despite its blood pressure lowering effects. Although ß-Pro7Ang III had no effect on aortic fibrosis, it significantly attenuated MCP-1 protein expression and superoxide levels when compared to both the non-diabetic and diabetic SHRs, to a similar extent as candesartan. In both the heart and vasculature, the effects of ß-Pro7Ang III in combination with candesartan were similar to those of ß-Pro7Ang III alone, and superior to candesartan alone. It was concluded that in hypertension with concomitant diabetes, AT2R stimulation with a novel ligand alone, or in combination with AT1R blockade, improved the cardiac and vascular structural changes that were strongly associated with inflammation and oxidative stress, independent of blood pressure regulation.


Assuntos
Diabetes Mellitus , Hipertensão , Animais , Ratos , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Ratos Endogâmicos SHR , Receptor Tipo 1 de Angiotensina/metabolismo , Superóxidos , Cardiotônicos
9.
Br J Pharmacol ; 2023 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-37095602

RESUMO

Heart failure remains a leading cause of morbidity and mortality worldwide. Current treatment for patients with heart failure include drugs targeting G protein-coupled receptors such as ß-adrenoceptor antagonists (ß-blockers) and angiotensin II type 1 receptor antagonists (or angiotensin II receptor blockers). However, many patients progress to advanced heart failure with persistent symptoms, despite treatment with available therapeutics that have been shown to reduce mortality and mortality. GPCR targets currently being explored for the development of novel heart failure therapeutics include adenosine receptor, formyl peptide receptor, relaxin/insulin-like family peptide receptor, vasopressin receptor, endothelin receptor and the glucagon-like peptide 1 receptor. Many GPCR drug candidates are limited by insufficient efficacy and/or dose-limiting unwanted effects. Understanding the current challenges hindering successful clinical translation and the potential to overcome existing limitations will facilitate the future development of novel heart failure therapeutics.

10.
Colloids Surf B Biointerfaces ; 199: 111557, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33434880

RESUMO

Diabetic infection is a long-term complication difficult to cure. The skin of diabetic patients is prone to damage, the healing is slow after the injury, and the wound occurs repeatedly. Therefore, there is an urgent need to develop an effective method for treating diabetes wounds. In this study, we used the electrospinning technique to load Huangbai Liniment (Compound Phellodendron Liquid, CPL) into Silk fibroin (SF) /poly-(L-lactide-co-caprolactone) (PLCL) to prepare the nanofiber membrane (SP/CPL) to treat the diabetic wound. The morphology and structure of the nanofibers were observed by scanning electron microscope (SEM). The SEM results indicate the smooth and bead free fibers and the diameter of the fiber decreased with increasing drug concentration. The release profile indicates the sustained release of the drug. Moreover, the drug-loaded nanofibers showed inhibitory effects for S.aureus and E.coli. Furthermore, in vitro cell culture studies showed the increased proliferation and adhesion of NIH-3T3 cells on the drug-containing nanofiber membrane. Animal experiments showed that the nanofiber membrane loaded with CPL increases the expression of the TGF-ß signaling pathway and collagen during wound healing, inhibits the expression of pro-inflammatory factors, and thus effectively promotes wound healing in diabetic mice. Therefore, the SP/CPL nanofiber scaffold with CPL loading is a potential candidate for diabetic wound dressings and tissue engineering.


Assuntos
Diabetes Mellitus Experimental , Fibroínas , Nanofibras , Animais , Caproatos , Diabetes Mellitus Experimental/tratamento farmacológico , Dioxanos , Medicamentos de Ervas Chinesas , Humanos , Lactonas , Linimentos , Camundongos , Poliésteres , Seda , Alicerces Teciduais , Cicatrização
11.
Nat Commun ; 12(1): 6157, 2021 10 25.
Artigo em Inglês | MEDLINE | ID: mdl-34697316

RESUMO

The pathogen Staphylococcus aureus can readily develop antibiotic resistance and evade the human immune system, which is associated with reduced levels of neutrophil recruitment. Here, we present a class of antibacterial peptides with potential to act both as antibiotics and as neutrophil chemoattractants. The compounds, which we term 'antibiotic-chemoattractants', consist of a formylated peptide (known to act as chemoattractant for neutrophil recruitment) that is covalently linked to the antibiotic vancomycin (known to bind to the bacterial cell wall). We use a combination of in vitro assays, cellular assays, infection-on-a-chip and in vivo mouse models to show that the compounds improve the recruitment, engulfment and killing of S. aureus by neutrophils. Furthermore, optimizing the formyl peptide sequence can enhance neutrophil activity through differential activation of formyl peptide receptors. Thus, we propose antibiotic-chemoattractants as an alternate approach for antibiotic development.


Assuntos
Antibacterianos/farmacologia , Fatores Quimiotáticos/farmacologia , Neutrófilos/efeitos dos fármacos , Staphylococcus aureus/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antibacterianos/química , Antibacterianos/uso terapêutico , Carga Bacteriana/efeitos dos fármacos , Fatores Quimiotáticos/química , Fatores Quimiotáticos/uso terapêutico , Farmacorresistência Bacteriana/efeitos dos fármacos , Imunoterapia , Camundongos , Neutrófilos/imunologia , Neutrófilos/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Fagocitose/efeitos dos fármacos , Receptores de Formil Peptídeo/metabolismo , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia , Infecções Estafilocócicas/terapia , Vancomicina/química , Vancomicina/farmacologia
12.
Front Pharmacol ; 11: 727, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32508651

RESUMO

Diabetes is associated with an increased mortality risk due to cardiovascular complications. Hyperglycemia-induced oxidative stress underlies these complications, leading to an impairment in endogenous nitric oxide (NO•) generation, together with reductions in NO• bioavailability and NO• responsiveness in the vasculature, platelets and myocardium. The latter impairment of responsiveness to NO•, termed NO• resistance, compromises the ability of traditional NO•-based therapeutics to improve hemodynamic status during diabetes-associated cardiovascular emergencies, such as acute myocardial infarction. Whilst a number of agents can ameliorate (e.g. angiotensin converting enzyme [ACE] inhibitors, perhexiline, statins and insulin) or circumvent (e.g. nitrite and sGC activators) NO• resistance, nitroxyl (HNO) donors offer a novel opportunity to circumvent NO• resistance in diabetes. With a suite of vasoprotective properties and an ability to enhance cardiac inotropic and lusitropic responses, coupled with preserved efficacy in the setting of oxidative stress, HNO donors have intact therapeutic potential in the face of diminished NO• signaling. This review explores the major mechanisms by which hyperglycemia-induced oxidative stress drives NO• resistance, and the therapeutic potential of HNO donors to circumvent this to treat cardiovascular complications in type 2 diabetes mellitus.

13.
Br J Pharmacol ; 177(1): 217-233, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31479151

RESUMO

BACKGROUND AND PURPOSE: Endothelium-derived vasoconstriction is a hallmark of vascular dysfunction in hypertension. In some cases, an overproduction of endothelium-derived prostacyclin (PGI2 ) can cause contraction rather than relaxation. Relaxin is well known for its vasoprotective actions, but the possibility that this peptide could also reverse endothelium-derived vasoconstriction has never been investigated. We tested the hypothesis that short-term relaxin treatment mitigates endothelium-derived vasoconstriction in spontaneously hypertensive rats (SHR). EXPERIMENTAL APPROACH: Male Wistar Kyoto rats (WKY) and SHR were subcutaneously infused with either vehicle (20 mmol·L-1 sodium acetate) or relaxin (13.3 µg·kg-1 ·hr-1 ) using osmotic minipumps for 3 days. Vascular reactivity to the endothelium-dependent agonist ACh was assessed in vitro by wire myography. Quantitative PCR and LC-MS were used to identify changes in gene expression of prostanoid pathways and PG production, respectively. KEY RESULTS: Relaxin treatment ameliorated hypertension-induced endothelial dysfunction by increasing NO-dependent relaxation and reducing endothelium-dependent contraction. Notably, short-term relaxin treatment up-regulated mesenteric PGI2 receptor (IP) expression, permitting PGI2 -IP-mediated vasorelaxation. In the aorta, reversal of contraction was accompanied by suppression of the hypertension-induced increase in prostanoid-producing enzymes and reduction in PGI2 -evoked contractions. CONCLUSIONS AND IMPLICATIONS: Relaxin has region-dependent vasoprotective actions in hypertension. Specifically, relaxin has distinct effects on endothelium-derived contracting factors and their associated vasoconstrictor pathways in mesenteric arteries and the aorta. Taken together, these observations reveal the potential of relaxin as a new therapeutic agent for vascular disorders that are associated with endothelium-derived vasoconstriction including hypertension.


Assuntos
Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Hipertensão/tratamento farmacológico , Hipertensão/metabolismo , Relaxina/uso terapêutico , Vasoconstrição/efeitos dos fármacos , Animais , Relação Dose-Resposta a Droga , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Artérias Mesentéricas/metabolismo , Ratos , Ratos Endogâmicos SHR , Ratos Endogâmicos WKY , Relaxina/farmacologia , Vasoconstrição/fisiologia
14.
Antioxid Redox Signal ; 9(1): 101-13, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17115889

RESUMO

The objective was a comprehensive investigation of the mechanisms and sites of resveratrol cardioprotection during and following ischemia-reperfusion (I-R) injury, and to determine whether direct preservation of cardiomyocytes is an important site of cardioprotection. We now provide the first definitive evidence that resveratrol specifically protects cardiomyocytes from I-R injury via a combination of suppression of superoxide levels and activation of potassium channels. This protection is apparent whether resveratrol is present for the full duration of the insult or only on recovery. In addition, resveratrol improved postischemic recovery of left ventricular contractile function, attenuated myocardial injury, and increased myocardial activation of the survival kinase Akt in the intact heart. Furthermore, resveratrol elicited direct concentration-dependent protective actions on the vasculature (vasorelaxation, superoxide suppression) and enhanced endothelium-dependent vasodilatation. Resveratrol thus targets a number of consequences of myocardial I-R, including release of reactive oxygen species, loss of recovery of contractile function, and impaired endothelium-dependent vasodilatation. Previous evidence indicates that resveratrol elicits potent preconditioning in the heart. Given that myocardial ischemic events are often unpredictable in humans, the findings that resveratrol protection is also evident when administered during and/or after the insult adds new dimensions to the clinical potential of resveratrol.


Assuntos
Antioxidantes/farmacologia , Coração/efeitos dos fármacos , Miócitos Cardíacos/efeitos dos fármacos , Traumatismo por Reperfusão/prevenção & controle , Estilbenos/farmacologia , Animais , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/fisiologia , Relação Dose-Resposta a Droga , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Humanos , Hipóxia , Técnicas In Vitro , Masculino , Ratos , Ratos Sprague-Dawley , Resveratrol
15.
Curr Drug Targets ; 18(15): 1689-1711, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-26424394

RESUMO

BACKGROUND: Early reperfusion of the blocked vessel is critical to restore the blood flow to the ischemic myocardium to salvage myocardial tissue and improve clinical outcome. This reperfusion strategy after a period of ischemia, however, may elicit further myocardial damage named myocardial reperfusion injury. The manifestations of reperfusion injury include arrhythmias, myocardial stunning and micro-vascular dysfunction, in addition to significant cardiomyocyte death. It is suggested that an overproduction of reactive oxygen species, intracellular calcium overload and inflammatory cell infiltration are the most important features of myocardial ischemia-reperfusion injury. OBJECTIVE: In this review, various pharmacological interventions to treat myocardial reperfusion injury including the antioxidant flavonols, hydrogen sulfide, adenosine, opioids, incretin-based therapies and cyclosporin A which targets the mitochondrial permeability transition pore are discussed. CONCLUSION: The processes involved in reperfusion injury might provide targets for improved outcomes after myocardial infarction but thus far that aim has not been met in the clinic.


Assuntos
Cardiotônicos/uso terapêutico , Isquemia Miocárdica/prevenção & controle , Traumatismo por Reperfusão Miocárdica/prevenção & controle , Animais , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Cálcio/metabolismo , Cardiotônicos/farmacologia , Ciclosporina/farmacologia , Ciclosporina/uso terapêutico , Humanos , Proteínas de Transporte da Membrana Mitocondrial/efeitos dos fármacos , Poro de Transição de Permeabilidade Mitocondrial , Isquemia Miocárdica/metabolismo , Traumatismo por Reperfusão Miocárdica/metabolismo , Espécies Reativas de Oxigênio/metabolismo
16.
Sci Rep ; 7: 39604, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28067255

RESUMO

Serelaxin prevents endothelial dysfunction in the mouse aorta ex vivo and inhibits apoptosis in cardiomyocytes under acute hyperglycaemia. Less is known about the effects of serelaxin in an in vivo mouse model of diabetes. Therefore, we tested the hypothesis in streptozotocin (STZ)-treated mice that serelaxin is able to reverse diabetes-induced vascular dysfunction and cardiac remodelling. Mice were divided into citrate buffer + placebo, STZ + placebo and STZ + serelaxin (0.5 mg/kg/d, 2 weeks) groups. After 12 weeks of diabetes, sensitivity to the endothelium-dependent agonist acetylcholine (ACh) was reduced in the mesenteric artery. This was accompanied by an enhanced vasoconstrictor prostanoid contribution and a decrease in endothelium-derived hyperpolarisation (EDH)-mediated relaxation. Serelaxin restored endothelial function by increasing nitric oxide (NO)-mediated relaxation but not EDH. It also normalised the contribution of vasoconstrictor prostanoids to endothelial dysfunction and suppressed diabetes-induced hyper-responsiveness of the mesenteric artery to angiotensin II. Similarly, diabetes reduced ACh-evoked NO-mediated relaxation in the aorta which was reversed by serelaxin. In the left ventricle, diabetes promoted apoptosis, hypertrophy and fibrosis; serelaxin treatment reversed this ventricular apoptosis and hypertrophy, but had no effect on fibrosis. In summary, serelaxin reversed diabetes-induced endothelial dysfunction by enhancing NO-mediated relaxation in the mouse vasculature and attenuating left ventricular hypertrophy and apoptosis.


Assuntos
Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Tipo 1/complicações , Hipertrofia Ventricular Esquerda/tratamento farmacológico , Relaxina/administração & dosagem , Animais , Aorta Abdominal/efeitos dos fármacos , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Hipertrofia Ventricular Esquerda/complicações , Masculino , Artérias Mesentéricas/efeitos dos fármacos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/efeitos dos fármacos , Proteínas Recombinantes/administração & dosagem , Estreptozocina
17.
Circ Heart Fail ; 8(3): 572-81, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25737497

RESUMO

BACKGROUND: Nitroxyl (HNO), a redox congener of nitric oxide (NO·), is a novel regulator of cardiovascular function, combining concomitant positive inotropic, lusitropic, and vasodilator properties. Moreover, HNO exhibits myocardial antihypertrophic and superoxide-suppressing actions. Despite these favorable actions, the impact of chronic HNO administration has yet to be reported in the context of cardiomyopathy. Diabetic cardiomyopathy is characterized by early diastolic dysfunction and adverse left ventricular (LV) structural remodeling, with LV superoxide generation playing a major causal role. We tested the hypothesis that the HNO donor 1-nitrosocyclohexylacetate (1-NCA) limits cardiomyocyte hypertrophy and LV diastolic dysfunction in a mouse model of diabetes mellitus in vivo. METHODS AND RESULTS: Diabetes mellitus was induced in male FVB/N mice using streptozotocin. After 4 weeks, diabetic and nondiabetic mice were allocated to 1-NCA therapy (83 mg/kg per day IP) or vehicle and followed up for a further 4 weeks. Diabetes mellitus-induced LV diastolic dysfunction was evident on echocardiography-derived E and A wave velocities, E:A ratio, deceleration, and isovolumic relaxation times; LV systolic function was preserved. Increased LV cardiomyocyte size, hypertrophic and profibrotic gene expression, and upregulation of LV superoxide were also evident. These characteristics of diabetic cardiomyopathy were largely prevented by 1-NCA treatment. Selectivity of 1-NCA as an HNO donor was demonstrated by sensitivity of acute 1-NCA to l-cysteine but not to hydroxocobalamin in the normal rat heart ex vivo. CONCLUSIONS: Our studies provide the first evidence that HNO donors may represent a promising strategy for treatment of diabetic cardiomyopathy and implies therapeutic efficacy in settings of chronic heart failure.


Assuntos
Acetatos/farmacologia , Diabetes Mellitus Experimental/tratamento farmacológico , Cardiomiopatias Diabéticas/prevenção & controle , Doadores de Óxido Nítrico/farmacologia , Compostos Nitrosos/farmacologia , Disfunção Ventricular Esquerda/prevenção & controle , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Vasos Coronários/efeitos dos fármacos , Vasos Coronários/fisiopatologia , Diabetes Mellitus Experimental/diagnóstico , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/fisiopatologia , Cardiomiopatias Diabéticas/diagnóstico , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/fisiopatologia , Diástole , Relação Dose-Resposta a Droga , Hipertrofia Ventricular Esquerda/fisiopatologia , Hipertrofia Ventricular Esquerda/prevenção & controle , Masculino , Camundongos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Superóxidos/metabolismo , Vasodilatação/efeitos dos fármacos , Disfunção Ventricular Esquerda/diagnóstico , Disfunção Ventricular Esquerda/metabolismo , Disfunção Ventricular Esquerda/fisiopatologia , Remodelação Ventricular/efeitos dos fármacos
18.
Pharmacol Ther ; 148: 47-65, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25460034

RESUMO

Myocardial infarction (MI) and its resultant heart failure remains a major cause of death in the world. The current treatments for patients with MI are revascularization with thrombolytic agents or interventional procedures. These treatments have focused on restoring blood flow to the ischemic tissue to prevent tissue necrosis and preserve organ function. The restoration of blood flow after a period of ischemia, however, may elicit further myocardial damage, called reperfusion injury. Pharmacological interventions, such as antioxidant and Ca(2+) channel blockers, have shown premises in experimental settings; however, clinical studies have shown limited success. Thus, there is a need for the development of novel therapies to treat reperfusion injury. The therapeutic potential of glucocorticoid-regulated anti-inflammatory mediator annexin-A1 (ANX-A1) has recently been recognized in a range of systemic inflammatory disorders. ANX-A1 binds to and activates the family of formyl peptide receptors (G protein-coupled receptor family) to inhibit neutrophil activation, migration and infiltration. Until recently, studies on the cardioprotective actions of ANX-A1 and its peptide mimetics (Ac2-26, CGEN-855A) have largely focused on its anti-inflammatory effects as a mechanism of preserving myocardial viability following I-R injury. Our laboratory provided the first evidence of the direct protective action of ANX-A1 on myocardium, independent of inflammatory cells in vitro. We now review the potential for ANX-A1 based therapeutics to be seen as a "triple shield" therapy against myocardial I-R injury, limiting neutrophil infiltration and preserving both cardiomyocyte viability and contractile function. This novel therapy may thus represent a valuable clinical approach to improve outcome after MI.


Assuntos
Anexina A1/metabolismo , Infarto do Miocárdio/metabolismo , Animais , Anexina A1/farmacologia , Anexina A1/uso terapêutico , Cardiotônicos/farmacologia , Cardiotônicos/uso terapêutico , Humanos , Infarto do Miocárdio/tratamento farmacológico , Receptores de Formil Peptídeo/metabolismo
19.
Free Radic Biol Med ; 87: 137-47, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25937176

RESUMO

Diabetes-induced cardiac complications include left ventricular (LV) dysfunction and heart failure. We previously demonstrated that LV phosphoinositide 3-kinase p110α (PI3K) protects the heart against diabetic cardiomyopathy, associated with reduced NADPH oxidase expression and activity. Conversely, in dominant negative PI3K(p110α) transgenic mice (dnPI3K), reduced cardiac PI3K signaling exaggerated diabetes-induced cardiomyopathy, associated with upregulated NADPH oxidase. The goal was to examine whether chronic supplementation with the antioxidant coenzyme Q(10) (CoQ(10)) could attenuate LV superoxide and diabetic cardiomyopathy in a setting of impaired PI3K signaling. Diabetes was induced in 6-week-old nontransgenic and dnPI3K male mice via streptozotocin. After 4 weeks of diabetes, CoQ(10) supplementation commenced (10 mg/kg ip, 3 times/week, 8 weeks). At study end (12 weeks of diabetes), markers of LV function, cardiomyocyte hypertrophy, collagen deposition, NADPH oxidase, oxidative stress (3-nitrotyrosine), and concentrations of CoQ(9) and CoQ(10) were determined. LV NADPH oxidase (Nox2 gene expression and activity, and lucigenin-enhanced chemiluminescence), as well as oxidative stress, were increased by diabetes, exaggerated in diabetic dnPI3K mice, and attenuated by CoQ(10). Diabetes-induced LV diastolic dysfunction (prolonged deceleration time, elevated end-diastolic pressure, impaired E/A ratio), cardiomyocyte hypertrophy and fibrosis, expression of atrial natriuretic peptide, connective tissue growth factor, and ß-myosin heavy chain were all attenuated by CoQ(10). Chronic CoQ(10) supplementation attenuates aspects of diabetic cardiomyopathy, even in a setting of reduced cardiac PI3K protective signaling. Given that CoQ(10) supplementation has been suggested to have positive outcomes in heart failure patients, chronic CoQ(10) supplementation may be an attractive adjunct therapy for diabetic heart failure.


Assuntos
Classe I de Fosfatidilinositol 3-Quinases/biossíntese , Cardiomiopatias Diabéticas/tratamento farmacológico , Estresse Oxidativo/efeitos dos fármacos , Ubiquinona/análogos & derivados , Disfunção Ventricular Esquerda/tratamento farmacológico , Animais , Antioxidantes/administração & dosagem , Classe I de Fosfatidilinositol 3-Quinases/genética , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/genética , Cardiomiopatias Diabéticas/genética , Cardiomiopatias Diabéticas/patologia , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Transgênicos , NADPH Oxidases/biossíntese , NADPH Oxidases/genética , Fosfatidilinositol 3-Quinases/genética , Transdução de Sinais/efeitos dos fármacos , Ubiquinona/administração & dosagem , Ubiquinona/metabolismo , Disfunção Ventricular Esquerda/genética , Disfunção Ventricular Esquerda/patologia
20.
Br J Pharmacol ; 171(7): 1722-34, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24372173

RESUMO

BACKGROUND AND PURPOSE: The NO redox sibling nitroxyl (HNO) elicits soluble guanylyl cyclase (sGC)-dependent vasodilatation. HNO has high reactivity with thiols, which is attributed with HNO-enhanced left ventricular (LV) function. Here, we tested the hypothesis that the concomitant vasodilatation and inotropic actions induced by a HNO donor, Angeli's salt (sodium trioxodinitrate), were sGC-dependent and sGC-independent respectively. EXPERIMENTAL APPROACH: Haemodynamic responses to Angeli's salt (10 pmol-10 µmol), alone and in the presence of scavengers of HNO (L-cysteine, 4 mM) or of NO [hydroxocobalamin (HXC), 100 µM] or a selective inhibitor of sGC [1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ), 10 µM], a CGRP receptor antagonist (CGRP8-37 , 0.1 µM) or a blocker of voltage-dependent potassium channels [4-aminopyridine (4-AP), 1 mM] were determined in isolated hearts from male rats. KEY RESULTS: Angeli's salt elicited concomitant, dose-dependent increases in coronary flow and LV systolic and diastolic function. Both L-cysteine and ODQ shifted (but did not abolish) the dose-response curve of each of these effects to the right, implying contributions from HNO and sGC in both the vasodilator and inotropic actions. In contrast, neither HXC, CGRP8-37 nor 4-AP affected these actions. CONCLUSIONS AND IMPLICATIONS: Both vasodilator and inotropic actions of the HNO donor Angeli's salt were mediated in part by sGC-dependent mechanisms, representing the first evidence that sGC contributes to the inotropic and lusitropic action of HNO in the intact heart. Thus, HNO acutely enhances LV contraction and relaxation, while concomitantly unloading the heart, potentially beneficial actions in failing hearts.


Assuntos
Cardiotônicos/farmacologia , Vasos Coronários/efeitos dos fármacos , Guanilato Ciclase/metabolismo , Contração Miocárdica/efeitos dos fármacos , Nitritos/farmacologia , Óxidos de Nitrogênio/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Vasodilatação/efeitos dos fármacos , Vasodilatadores/farmacologia , Função Ventricular Esquerda/efeitos dos fármacos , Animais , Vasos Coronários/enzimologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Sequestradores de Radicais Livres/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Técnicas In Vitro , Masculino , Miocárdio/enzimologia , Bloqueadores dos Canais de Potássio/farmacologia , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Guanilil Ciclase Solúvel , Fatores de Tempo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA