Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Respir Crit Care Med ; 210(5): 581-592, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38984912

RESUMEN

Pulmonary arterial hypertension (PAH) is a complex fatal condition that requires aggressive treatment with close monitoring. Significant progress has been made over the last three decades in the treatment of PAH, but, despite this progress, survival has remained unacceptably low. In the quest to improve survival, therapeutic interventions play a central role. In the last few years, there have been remarkable attempts to identify novel treatments. Finally, we have had a breakthrough with the discovery of the fourth treatment pathway in PAH. Activin signaling inhibition distinguishes itself as a potential antiproliferative intervention as opposed to the traditional therapies, which mediate their effect primarily by vasodilatation. With this novel treatment pathway, we stand at an important milestone with an exciting future ahead and the natural question of when to use an activin signaling inhibitor for the treatment of PAH. In this state-of-the-art review, we focus on the placement of this novel agent in the PAH treatment paradigm, based on the available evidence, with special focus on the U.S. patient population. This review also provides an expert opinion of the current treatment algorithm in important subgroups of patients with comorbidities from the U.S. perspective.


Asunto(s)
Hipertensión Arterial Pulmonar , Humanos , Estados Unidos , Hipertensión Arterial Pulmonar/tratamiento farmacológico , Hipertensión Arterial Pulmonar/fisiopatología , Hipertensión Arterial Pulmonar/terapia , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/fisiopatología , Hipertensión Pulmonar/terapia , Antihipertensivos/uso terapéutico , Activinas
2.
N Engl J Med ; 384(4): 325-334, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33440084

RESUMEN

BACKGROUND: No therapies are currently approved for the treatment of pulmonary hypertension in patients with interstitial lung disease. The safety and efficacy of inhaled treprostinil for patients with this condition are unclear. METHODS: We enrolled patients with interstitial lung disease and pulmonary hypertension (documented by right heart catheterization) in a multicenter, randomized, double-blind, placebo-controlled, 16-week trial. Patients were assigned in a 1:1 ratio to receive inhaled treprostinil, administered by means of an ultrasonic, pulsed-delivery nebulizer in up to 12 breaths (total, 72 µg) four times daily, or placebo. The primary efficacy end point was the difference between the two groups in the change in peak 6-minute walk distance from baseline to week 16. Secondary end points included the change in N-terminal pro-B-type natriuretic peptide (NT-proBNP) level at week 16 and the time to clinical worsening. RESULTS: A total of 326 patients underwent randomization, with 163 assigned to inhaled treprostinil and 163 to placebo. Baseline characteristics were similar in the two groups. At week 16, the least-squares mean difference between the treprostinil group and the placebo group in the change from baseline in the 6-minute walk distance was 31.12 m (95% confidence interval [CI], 16.85 to 45.39; P<0.001). There was a reduction of 15% in NT-proBNP levels from baseline with inhaled treprostinil as compared with an increase of 46% with placebo (treatment ratio, 0.58; 95% CI, 0.47 to 0.72; P<0.001). Clinical worsening occurred in 37 patients (22.7%) in the treprostinil group as compared with 54 patients (33.1%) in the placebo group (hazard ratio, 0.61; 95% CI, 0.40 to 0.92; P = 0.04 by the log-rank test). The most frequently reported adverse events were cough, headache, dyspnea, dizziness, nausea, fatigue, and diarrhea. CONCLUSIONS: In patients with pulmonary hypertension due to interstitial lung disease, inhaled treprostinil improved exercise capacity from baseline, assessed with the use of a 6-minute walk test, as compared with placebo. (Funded by United Therapeutics; INCREASE ClinicalTrials.gov number, NCT02630316.).


Asunto(s)
Antihipertensivos/uso terapéutico , Epoprostenol/análogos & derivados , Hipertensión Pulmonar/tratamiento farmacológico , Enfermedades Pulmonares Intersticiales/complicaciones , Prueba de Paso , Administración por Inhalación , Adulto , Anciano , Anciano de 80 o más Años , Antihipertensivos/efectos adversos , Método Doble Ciego , Epoprostenol/efectos adversos , Epoprostenol/uso terapéutico , Tolerancia al Ejercicio/efectos de los fármacos , Femenino , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/fisiopatología , Análisis de los Mínimos Cuadrados , Masculino , Persona de Mediana Edad , Calidad de Vida
3.
Eur Respir J ; 63(6)2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38811045

RESUMEN

BACKGROUND: Pulmonary hypertension (PH) accompanying COPD (PH-COPD) is associated with worse outcomes than COPD alone. There are currently no approved therapies to treat PH-COPD. The PERFECT study (ClinicalTrials.gov: NCT03496623) evaluated the safety and efficacy of inhaled treprostinil (iTRE) in this patient population. METHODS: Patients with PH-COPD (mean pulmonary arterial pressure ≥30 mmHg and pulmonary vascular resistance ≥4 WU) were enrolled in a multicentre, randomised (1:1), double-blind, placebo-controlled, 12-week, crossover study. A contingent parallel design was also prespecified and implemented, based on a blinded interim analysis of missing data. Patients received treatment with iTRE up to 12 breaths (72 µg) 4 times daily or placebo. The primary efficacy end-point was change in peak 6-min walk distance (6MWD) at week 12. RESULTS: In total, 76 patients were randomised, 64 in the original crossover design and 12 in the contingent parallel design; 66 patients received iTRE and 58 received placebo. The study was terminated early at the recommendation of the data and safety monitoring committee based on the totality of evidence that iTRE increased the risk of serious adverse events and suggestive evidence of an increased risk of mortality. The change in 6MWD was numerically worse with iTRE exposure than with placebo exposure. CONCLUSIONS: The risk-benefit observations associated with iTRE in patients with PH-COPD did not support continuation of the PERFECT study. The results of this study do not support iTRE as a viable treatment option in patients with PH-COPD.


Asunto(s)
Antihipertensivos , Estudios Cruzados , Epoprostenol , Hipertensión Pulmonar , Enfermedad Pulmonar Obstructiva Crónica , Prueba de Paso , Humanos , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Epoprostenol/análogos & derivados , Epoprostenol/administración & dosificación , Epoprostenol/uso terapéutico , Femenino , Masculino , Hipertensión Pulmonar/tratamiento farmacológico , Administración por Inhalación , Anciano , Persona de Mediana Edad , Método Doble Ciego , Antihipertensivos/administración & dosificación , Antihipertensivos/uso terapéutico , Resultado del Tratamiento
4.
Am J Respir Crit Care Med ; 207(6): 740-756, 2023 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-36343281

RESUMEN

Rationale: Inflammation drives pulmonary arterial hypertension (PAH). Gut dysbiosis causes immune dysregulation and systemic inflammation by altering circulating microbial metabolites; however, little is known about gut dysbiosis and microbial metabolites in PAH. Objectives: To characterize the gut microbiome and microbial metabolites in patients with PAH. Methods: We performed 16S ribosomal RNA gene and shotgun metagenomics sequencing on stool from patients with PAH, family control subjects, and healthy control subjects. We measured markers of inflammation, gut permeability, and microbial metabolites in plasma from patients with PAH, family control subjects, and healthy control subjects. Measurements and Main Results: The gut microbiome was less diverse in patients with PAH. Shannon diversity index correlated with measures of pulmonary vascular disease but not with right ventricular function. Patients with PAH had a distinct gut microbial signature at the phylogenetic level, with fewer copies of gut microbial genes that produce antiinflammatory short-chain fatty acids (SCFAs) and secondary bile acids and lower relative abundances of species encoding these genes. Consistent with the gut microbial changes, patients with PAH had relatively lower plasma concentrations of SCFAs and secondary bile acids. Patients with PAH also had enrichment of species with the microbial genes that encoded the proinflammatory microbial metabolite trimethylamine. The changes in the gut microbiome and circulating microbial metabolites between patients with PAH and family control subjects were not as substantial as the differences between patients with PAH and healthy control subjects. Conclusions: Patients with PAH have proinflammatory gut dysbiosis, in which lower circulating SCFAs and secondary bile acids may facilitate pulmonary vascular disease. These findings support investigating modulation of the gut microbiome as a potential treatment for PAH.


Asunto(s)
Microbioma Gastrointestinal , Hipertensión Arterial Pulmonar , Enfermedades Vasculares , Humanos , Microbioma Gastrointestinal/genética , Disbiosis , Filogenia , Hipertensión Pulmonar Primaria Familiar , Inflamación , Ácidos y Sales Biliares
5.
Circulation ; 146(7): e73-e88, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35862198

RESUMEN

This science advisory focuses on the need to better understand the epidemiology, pathophysiology, and treatment of pulmonary hypertension in patients with heart failure with preserved ejection fraction. This clinical phenotype is important because it is common, is strongly associated with adverse outcomes, and lacks evidence-based therapies. Our goal is to clarify key knowledge gaps in pulmonary hypertension attributable to heart failure with preserved ejection fraction and to suggest specific, actionable scientific directions for addressing such gaps. Areas in need of additional investigation include refined disease definitions and interpretation of hemodynamics, as well as greater insights into noncardiac contributors to pulmonary hypertension risk, optimized animal models, and further molecular studies in patients with combined precapillary and postcapillary pulmonary hypertension. We highlight translational approaches that may provide important biological insight into pathophysiology and reveal new therapeutic targets. Last, we discuss the current and future landscape of potential therapies for patients with heart failure with preserved ejection fraction and pulmonary vascular dysfunction, including considerations of precision medicine, novel trial design, and device-based therapies, among other considerations. This science advisory provides a synthesis of important knowledge gaps, culminating in a collection of specific research priorities that we argue warrant investment from the scientific community.


Asunto(s)
Insuficiencia Cardíaca , Hipertensión Pulmonar , American Heart Association , Animales , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/terapia , Volumen Sistólico/fisiología , Función Ventricular Izquierda
6.
Eur Respir J ; 61(6)2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37080567

RESUMEN

INTRODUCTION: The 16-week randomised, placebo-controlled INCREASE trial (RCT) met its primary end-point by improving 6-min walk distance (6MWD) in patients receiving inhaled treprostinil for pulmonary hypertension due to interstitial lung disease (PH-ILD). The open-label extension (OLE) evaluated long-term effects of inhaled treprostinil in PH-ILD. METHODS: Of 258 eligible patients, 242 enrolled in the INCREASE OLE and received inhaled treprostinil. Assessments included 6MWD, pulmonary function testing, N-terminal pro-brain natriuretic peptide (NT-proBNP), quality of life and adverse events. Hospitalisations, exacerbations of underlying lung disease and death were recorded. RESULTS: At INCREASE OLE baseline, patients had a median age of 70 years and a mean 6MWD of 274.2 m; 52.1% were male. For the overall population, the mean 6MWD at week 52 was 279.1 m and the mean change from INCREASE RCT baseline was 3.5 m (22.1 m for the prior inhaled treprostinil arm and -19.5 m for the prior placebo arm); the median NT-proBNP decreased from 389 pg·mL-1 at RCT baseline to 359 pg·mL-1 at week 64; and the absolute (% predicted) mean forced vital capacity change from RCT baseline to week 64 was 51 mL (2.8%). Patients who received inhaled treprostinil versus placebo in the RCT had a 31% lower relative risk of exacerbation of underlying lung disease in the OLE (hazard ratio 0.69 (95% CI 0.49-0.97); p=0.03). Adverse events leading to drug discontinuation occurred in 54 (22.3%) patients. CONCLUSIONS: These results support the long-term safety and efficacy of inhaled treprostinil in patients with PH-ILD, and are consistent with the results observed in the INCREASE RCT.


Asunto(s)
Hipertensión Pulmonar , Enfermedades Pulmonares Intersticiales , Anciano , Femenino , Humanos , Masculino , Antihipertensivos/uso terapéutico , Epoprostenol , Hipertensión Pulmonar/complicaciones , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/inducido químicamente , Enfermedades Pulmonares Intersticiales/complicaciones , Enfermedades Pulmonares Intersticiales/tratamiento farmacológico , Calidad de Vida , Resultado del Tratamiento
7.
Curr Opin Pulm Med ; 27(5): 296-302, 2021 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-34175858

RESUMEN

PURPOSE OF REVIEW: Treatment options for Group 3 pulmonary hypertension, characterized as secondary to chronic hypoxia or lung disease, remain an elusive holy grail for physicians and patients alike. Despite increasing identification and investigation into this pulmonary vasculopathy group with the second-highest frequency and highest mortality, there are no therapeutic interventions that offer the significant improvements in morbidity and mortality comparable to those benefiting other pulmonary hypertension groups including pulmonary arterial hypertension. This review examines the data on available and emerging Group 3 pulmonary hypertension treatments. RECENT FINDINGS: Pulmonary vasodilators have yielded equivocal results in this patient population, although recent evidence shows modestly improved outcomes with inhaled treprostinil in interstitial lung disease-associated pulmonary hypertension. With pulmonary vasodilators providing limited benefit, emerging data support the right ventricle as a potential treatment target in Group 3 pulmonary hypertension. SUMMARY: Group 3 pulmonary hypertension is associated with significant morbidity and mortality. Pulmonary vasodilators offer only limited haemodynamic and exertional benefits, and lung transplantation remains the only cure for this deadly disease. The right ventricle may provide a novel intervention target.


Asunto(s)
Hipertensión Pulmonar , Trasplante de Pulmón , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Arteria Pulmonar , Circulación Pulmonar , Vasodilatadores/uso terapéutico
8.
Int J Mol Sci ; 21(19)2020 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-33019763

RESUMEN

The hexosamine biosynthetic pathway (HBP) converts glucose to uridine-diphosphate-N-acetylglucosamine, which, when added to serines or threonines, modulates protein function through protein O-GlcNAcylation. Glutamine-fructose-6-phosphate amidotransferase (GFAT) regulates HBP flux, and AMP-kinase phosphorylation of GFAT blunts GFAT activity and O-GlcNAcylation. While numerous studies demonstrate increased right ventricle (RV) glucose uptake in pulmonary arterial hypertension (PAH), the relationship between O-GlcNAcylation and RV function in PAH is unexplored. Therefore, we examined how colchicine-mediated AMP-kinase activation altered HBP intermediates, O-GlcNAcylation, mitochondrial function, and RV function in pulmonary artery-banded (PAB) and monocrotaline (MCT) rats. AMPK activation induced GFAT phosphorylation and reduced HBP intermediates and O-GlcNAcylation in MCT but not PAB rats. Reduced O-GlcNAcylation partially restored the RV metabolic signature and improved RV function in MCT rats. Proteomics revealed elevated expression of O-GlcNAcylated mitochondrial proteins in MCT RVs, which fractionation studies corroborated. Seahorse micropolarimetry analysis of H9c2 cardiomyocytes demonstrated colchicine improved mitochondrial function and reduced O-GlcNAcylation. Presence of diabetes in PAH, a condition of excess O-GlcNAcylation, reduced RV contractility when compared to nondiabetics. Furthermore, there was an inverse relationship between RV contractility and HgbA1C. Finally, RV biopsy specimens from PAH patients displayed increased O-GlcNAcylation. Thus, excess O-GlcNAcylation may contribute to metabolic derangements and RV dysfunction in PAH.


Asunto(s)
Diabetes Mellitus/metabolismo , Hipertrofia Ventricular Derecha/metabolismo , Mitocondrias/metabolismo , Procesamiento Proteico-Postraduccional , Disfunción Ventricular Derecha/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Acilación , Adulto , Anciano , Animales , Línea Celular , Estudios de Cohortes , Colchicina/farmacología , Diabetes Mellitus/diagnóstico por imagen , Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatología , Modelos Animales de Enfermedad , Ecocardiografía , Regulación de la Expresión Génica , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/genética , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/metabolismo , Hexosaminas/metabolismo , Humanos , Hipertrofia Ventricular Derecha/diagnóstico por imagen , Hipertrofia Ventricular Derecha/genética , Hipertrofia Ventricular Derecha/fisiopatología , Masculino , Metaboloma , Persona de Mediana Edad , Mitocondrias/efectos de los fármacos , Monocrotalina/administración & dosificación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Ratas , Ratas Sprague-Dawley , Disfunción Ventricular Derecha/diagnóstico por imagen , Disfunción Ventricular Derecha/genética , Disfunción Ventricular Derecha/fisiopatología
9.
Am J Physiol Heart Circ Physiol ; 317(5): H1093-H1101, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31490732

RESUMEN

Pulmonary arterial hypertension (PAH) is a fatal disease with a median survival of only 5-7 yr. PAH is characterized by remodeling of the pulmonary vasculature causing reduced pulmonary arterial compliance (PAC) and increased pulmonary vascular resistance (PVR), ultimately resulting in right ventricular failure and death. Better therapies for PAH will require a paradigm shift in our understanding of the early pathophysiology. PAC decreases before there is an increase in the PVR. Unfortunately, present treatment has little effect on PAC. The loss of compliance correlates with extracellular matrix remodeling and fibrosis in the pulmonary vessels, which have been linked to chronic perivascular inflammation and immune dysregulation. However, what initiates the perivascular inflammation and immune dysregulation in PAH is unclear. Alteration of the gut microbiota composition and function underlies the level of immunopathogenic involvement in several diseases, including atherosclerosis, obesity, diabetes mellitus, and depression, among others. In this review, we discuss evidence that raises the possibility of an etiologic role for changes in the gut and circulating microbiome in the initiation of perivascular inflammation in the early pathogenesis of PAH.


Asunto(s)
Presión Arterial , Bacterias/metabolismo , Microbioma Gastrointestinal , Mediadores de Inflamación/sangre , Intestinos/microbiología , Hipertensión Arterial Pulmonar/microbiología , Arteria Pulmonar/microbiología , Animales , Bacterias/inmunología , Disbiosis , Interacciones Huésped-Patógeno , Humanos , Mediadores de Inflamación/inmunología , Hipertensión Arterial Pulmonar/sangre , Hipertensión Arterial Pulmonar/inmunología , Hipertensión Arterial Pulmonar/fisiopatología , Arteria Pulmonar/inmunología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Factores de Riesgo , Transducción de Señal
11.
Am J Physiol Heart Circ Physiol ; 315(5): H1322-H1331, 2018 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-30141981

RESUMEN

Pulmonary arterial hypertension (PAH) is characterized by remodeling of the extracellular matrix (ECM) of the pulmonary arteries with increased collagen deposition, cross-linkage of collagen, and breakdown of elastic laminae. Extracellular matrix remodeling occurs due to an imbalance in the proteolytic enzymes, such as matrix metalloproteinases, elastases, and lysyl oxidases, and tissue inhibitor of matrix metalloproteinases, which, in turn, results from endothelial cell dysfunction, endothelial-to-mesenchymal transition, and inflammation. ECM remodeling and pulmonary vascular stiffness occur early in the disease process, before the onset of the increase in the intimal and medial thickness and pulmonary artery pressure, suggesting that the ECM is a cause rather than a consequence of distal pulmonary vascular remodeling. ECM remodeling and increased pulmonary arterial stiffness promote proliferation of pulmonary vascular cells (endothelial cells, smooth muscle cells, and adventitial fibroblasts) through mechanoactivation of various signaling pathways, including transcriptional cofactors YAP/TAZ, transforming growth factor-ß, transient receptor potential channels, Toll-like receptor, and NF-κB. Inhibition of ECM remodeling and mechanotransduction prevents and reverses experimental pulmonary hypertension. These data support a central role for ECM remodeling in the pathogenesis of the PAH, making it an attractive novel therapeutic target.


Asunto(s)
Presión Arterial , Matriz Extracelular/metabolismo , Hipertensión Pulmonar/metabolismo , Mecanotransducción Celular , Arteria Pulmonar/metabolismo , Remodelación Vascular , Rigidez Vascular , Animales , Antihipertensivos/uso terapéutico , Presión Arterial/efectos de los fármacos , Colágeno/metabolismo , Adaptabilidad , Matriz Extracelular/efectos de los fármacos , Matriz Extracelular/patología , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Mecanotransducción Celular/efectos de los fármacos , Terapia Molecular Dirigida , Arteria Pulmonar/efectos de los fármacos , Arteria Pulmonar/patología , Arteria Pulmonar/fisiopatología , Remodelación Vascular/efectos de los fármacos , Rigidez Vascular/efectos de los fármacos
13.
Am J Emerg Med ; 35(7): 1041.e5-1041.e6, 2017 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-28292545

RESUMEN

A 21year-old male presented to the emergency department with 6 h of atypical chest pain after suffering blunt chest trauma. His electrocardiogram revealed 1-1.5mm ST segment elevation in leads V1-V3 with reciprocal depressions in II, III, and aVF. Mid-anterior wall akinesis was observed on echocardiography associated with an estimated left ventricular ejection fraction of 40%. A left main coronary artery dissection was diagnosed and treated surgically with a bypass graft. Although rare, coronary dissections can be a catastrophic complication of chest trauma.


Asunto(s)
Disección Aórtica/diagnóstico por imagen , Dolor en el Pecho/diagnóstico por imagen , Aneurisma Coronario/diagnóstico por imagen , Angiografía Coronaria , Puente de Arteria Coronaria , Traumatismos Torácicos/fisiopatología , Heridas no Penetrantes/fisiopatología , Disección Aórtica/fisiopatología , Disección Aórtica/cirugía , Arritmias Cardíacas/fisiopatología , Aneurisma Coronario/fisiopatología , Aneurisma Coronario/cirugía , Ecocardiografía , Medicina de Emergencia , Humanos , Masculino , Traumatismos Torácicos/complicaciones , Traumatismos Torácicos/cirugía , Resultado del Tratamiento , Disfunción Ventricular Izquierda/diagnóstico por imagen , Heridas no Penetrantes/complicaciones , Heridas no Penetrantes/cirugía , Adulto Joven
15.
Curr Heart Fail Rep ; 13(2): 92-102, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26886381

RESUMEN

Pulmonary hypertension associated with left heart disease is the most common form of pulmonary hypertension. Although its pathophysiology remains incompletely understood, it is now well recognized that the presence of pulmonary hypertension is associated with a worse prognosis. Right ventricular failure has independent and additive prognostic value over pulmonary hypertension for adverse outcomes in left heart disease. Recently, several new terminologies have been introduced to better define and characterize the nature and severity of pulmonary hypertension. Several new treatment options including the use of pulmonary arterial hypertension specific therapies are being considered, but there is lack of evidence. Here, we review the recent advances in this field and summarize the diagnostic and therapeutic modalities of use in the management of pulmonary hypertension associated with left heart disease.


Asunto(s)
Insuficiencia Cardíaca/fisiopatología , Hipertensión Pulmonar/fisiopatología , Disfunción Ventricular Izquierda/fisiopatología , Disfunción Ventricular Derecha/fisiopatología , Cardiopatías/fisiopatología , Humanos , Hipertensión Pulmonar/etiología , Pronóstico
16.
J Clin Outcomes Manag ; 22(10): 443-454, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27158218

RESUMEN

OBJECTIVE: To present a review of cardiorenal syndrome type 1 (CRS1). METHODS: Review of the literature. RESULTS: Acute kidney injury occurs in approximately one-third of patients with acute decompensated heart failure (ADHF) and the resultant condition was named CRS1. A growing body of literature shows CRS1 patients are at high risk for poor outcomes, and thus there is an urgent need to understand the pathophysiology and subsequently develop effective treatments. In this review we discuss prevalence, proposed pathophysiology including hemodynamic and nonhemodynamic factors, prognosticating variables, data for different treatment strategies, and ongoing clinical trials and highlight questions and problems physicians will face moving forward with this common and challenging condition. CONCLUSION: Further research is needed to understand the pathophysiology of this complex clinical entity and to develop effective treatments.

17.
Circ Res ; 110(11): 1484-97, 2012 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-22511751

RESUMEN

RATIONALE: Pulmonary arterial hypertension (PAH) is a lethal syndrome characterized by pulmonary vascular obstruction caused, in part, by pulmonary artery smooth muscle cell (PASMC) hyperproliferation. Mitochondrial fragmentation and normoxic activation of hypoxia-inducible factor-1α (HIF-1α) have been observed in PAH PASMCs; however, their relationship and relevance to the development of PAH are unknown. Dynamin-related protein-1 (DRP1) is a GTPase that, when activated by kinases that phosphorylate serine 616, causes mitochondrial fission. It is, however, unknown whether mitochondrial fission is a prerequisite for proliferation. OBJECTIVE: We hypothesize that DRP1 activation is responsible for increased mitochondrial fission in PAH PASMCs and that DRP1 inhibition may slow proliferation and have therapeutic potential. METHODS AND RESULTS: Experiments were conducted using human control and PAH lungs (n=5) and PASMCs in culture. Parallel experiments were performed in rat lung sections and PASMCs and in rodent PAH models induced by the HIF-1α activator, cobalt, chronic hypoxia, and monocrotaline. HIF-1α activation in human PAH leads to mitochondrial fission by cyclin B1/CDK1-dependent phosphorylation of DRP1 at serine 616. In normal PASMCs, HIF-1α activation by CoCl(2) or desferrioxamine causes DRP1-mediated fission. HIF-1α inhibition reduces DRP1 activation, prevents fission, and reduces PASMC proliferation. Both the DRP1 inhibitor Mdivi-1 and siDRP1 prevent mitotic fission and arrest PAH PASMCs at the G2/M interphase. Mdivi-1 is antiproliferative in human PAH PASMCs and in rodent models. Mdivi-1 improves exercise capacity, right ventricular function, and hemodynamics in experimental PAH. CONCLUSIONS: DRP-1-mediated mitotic fission is a cell-cycle checkpoint that can be therapeutically targeted in hyperproliferative disorders such as PAH.


Asunto(s)
Proliferación Celular , Dinaminas/metabolismo , GTP Fosfohidrolasas/metabolismo , Hipertensión Pulmonar/enzimología , Proteínas Asociadas a Microtúbulos/metabolismo , Mitocondrias Musculares/enzimología , Proteínas Mitocondriales/metabolismo , Mitosis , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Animales , Antihipertensivos/farmacología , Proteína Quinasa CDC2/metabolismo , Estudios de Casos y Controles , Puntos de Control del Ciclo Celular , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cobalto , Ciclina B1/metabolismo , Modelos Animales de Enfermedad , Dinaminas/genética , Activación Enzimática , Hipertensión Pulmonar Primaria Familiar , GTP Fosfohidrolasas/genética , Terapia Genética/métodos , Glucólisis , Humanos , Hipertensión Pulmonar/etiología , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/terapia , Hipoxia/complicaciones , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Masculino , Proteínas Asociadas a Microtúbulos/genética , Mitocondrias Musculares/efectos de los fármacos , Mitocondrias Musculares/patología , Proteínas Mitocondriales/genética , Mitosis/efectos de los fármacos , Monocrotalina , Músculo Liso Vascular/efectos de los fármacos , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/patología , Fosforilación , Arteria Pulmonar/enzimología , Arteria Pulmonar/patología , Quinazolinonas/farmacología , Interferencia de ARN , Ratas , Ratas Sprague-Dawley , Serina , Factores de Tiempo , Transfección
19.
Curr Heart Fail Rep ; 11(4): 428-35, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25135469

RESUMEN

Pulmonary hypertension (PH) leading to right ventricular failure (RVF) is a common complication of left heart failure irrespective of the left ventricular ejection fraction. PH due to left heart disease is the most common cause of PH. The prevalence of PH and RVF in left heart failure varies depending on the patient population studied, the method used to diagnose PH, and the hemodynamic criteria used to define PH. Elevated left-sided filling pressure and functional mitral regurgitation are the two major determinants of PH in left heart failure. PH is associated with markers of disease severity, advanced symptoms, and worse long-term outcomes including heart failure hospitalization and mortality in left heart failure. RVF has independent, incremental prognostic value over PH for adverse outcomes in left heart failure. PH and RVF may be potential therapeutic targets in patients with left heart failure.


Asunto(s)
Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/epidemiología , Hipertensión Pulmonar/epidemiología , Hipertensión Pulmonar/etiología , Disfunción Ventricular Derecha/epidemiología , Disfunción Ventricular Derecha/etiología , Insuficiencia Cardíaca/fisiopatología , Hemodinámica , Humanos , Hipertensión Pulmonar/fisiopatología , Prevalencia , Pronóstico , Disfunción Ventricular Derecha/fisiopatología
20.
bioRxiv ; 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39229028

RESUMEN

Emerging data demonstrate systemic and local inflammation regulate right ventricular (RV) adaption in preclinical and human pulmonary arterial hypertension (PAH). Pathological RV inflammation is targetable as antagonism of glycoprotein-130 (GP130) signaling counteracts pathological microtubule remodeling and improves RV function in rodents. Microtubules control several aspects of cardiomyocyte biology including cellular and nuclear size/structure, t-tubule homeostasis, and the proper localization of connexin-43. The intestinal microbiome regulates systemic inflammation, but the impact of the gut microbiome on the GP130-microtubule axis in RV failure is unknown. Here, we examined how the anti-inflammatory bacteria, Lactobacillus , modulated cellular and physiological RV phenotypes in preclinical and clinical PAH. Lactobacillus supplementation restructured the gut micro/mycobiome, suppressed systemic inflammation, combatted pathological GP130-mediated RV cardiomyocyte microtubule remodeling, and augmented RV function in rodent PAH. Moreover, Lactobacillus was associated with superior RV adaption in human PAH. These data further support the hypothesis that inflammation negatively impacts RV adaption in PAH, and identify the gut microbiome as a potentially targetable regulator of RV function in PAH.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA