Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 69(4): 689-698.e7, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29429925

RESUMEN

Endothelial-to-mesenchymal transition (EndoMT) is a cellular process often initiated by the transforming growth factor ß (TGF-ß) family of ligands. Although required for normal heart valve development, deregulated EndoMT is linked to a wide range of pathological conditions. Here, we demonstrate that endothelial fatty acid oxidation (FAO) is a critical in vitro and in vivo regulator of EndoMT. We further show that this FAO-dependent metabolic regulation of EndoMT occurs through alterations in intracellular acetyl-CoA levels. Disruption of FAO via conditional deletion of endothelial carnitine palmitoyltransferase II (Cpt2E-KO) augments the magnitude of embryonic EndoMT, resulting in thickening of cardiac valves. Consistent with the known pathological effects of EndoMT, adult Cpt2E-KO mice demonstrate increased permeability in multiple vascular beds. Taken together, these results demonstrate that endothelial FAO is required to maintain endothelial cell fate and that therapeutic manipulation of endothelial metabolism could provide the basis for treating a growing number of EndoMT-linked pathological conditions.


Asunto(s)
Carnitina O-Palmitoiltransferasa/fisiología , Endotelio Vascular/metabolismo , Transición Epitelial-Mesenquimal , Ácidos Grasos/química , 3-Hidroxiacil-CoA Deshidrogenasas , Acetilcoenzima A/metabolismo , Acetil-CoA C-Aciltransferasa , Animales , Isomerasas de Doble Vínculo Carbono-Carbono , Células Cultivadas , Endotelio Vascular/citología , Enoil-CoA Hidratasa , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Oxidación-Reducción , Racemasas y Epimerasas , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
2.
J Immunol ; 205(4): 1125-1134, 2020 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-32601098

RESUMEN

NF-κB is a reduction-oxidation-sensitive transcription factor that plays a key role in regulating the immune response. In these studies, we intended to investigate the role of mitochondrial-derived reactive oxygen species in regulating NF-κB activation by studying transgenic mice that overexpress mitochondrial-targeted human catalase (mCAT). We treated wild-type (WT) and mCAT mice with intratracheal instillation of Escherichia coli LPS and found that mCAT mice had exaggerated NF-κB activation in the lungs, increased neutrophilic alveolitis, and greater lung inflammation/injury compared with WT mice. Additional studies using bone marrow chimeras revealed that this hyperinflammatory phenotype was mediated by immune/inflammatory cells. Mechanistic studies using bone marrow-derived macrophages (BMDMs) showed that LPS treatment induced a sustained increase in NF-κB activation and expression of NF-κB-dependent inflammatory mediators in mCAT BMDMs compared with WT BMDMs. Further investigations showed that cytoplasmic, but not mitochondrial, hydrogen peroxide levels were reduced in LPS-treated mCAT BMDMs. However, mCAT macrophages exhibited increased glycolytic and oxidative metabolism, coupled with increased ATP production and an increased intracellular NADH/NAD+ ratio compared with BMDMs from WT mice. Treatment of BMDMs with lactate increased the intracellular NADH/NAD+ ratio and upregulated NF-κB activation after LPS treatment, whereas treatment with a potent inhibitor of the mitochondrial pyruvate carrier (UK5099) decreased the NADH/NAD+ ratio and reduced NF-κB activation. Taken together, these findings point to an increased availability of reducing equivalents in the form of NADH as an important mechanism by which metabolic activity modulates inflammatory signaling through the NF-κB pathway.


Asunto(s)
Catalasa/metabolismo , Macrófagos/metabolismo , Mitocondrias/metabolismo , FN-kappa B/metabolismo , Neumonía/metabolismo , Animales , Médula Ósea/metabolismo , Pulmón/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , NAD/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/fisiología
3.
Crit Care Med ; 49(11): 1963-1973, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34495876

RESUMEN

Given the urgent need for coronavirus disease 2019 therapeutics, early in the pandemic the Accelerating Coronavirus Disease 2019 Therapeutic Interventions and Vaccines (ACTIV) public-private partnership rapidly designed a unique therapeutic agent intake and assessment process for candidate treatments of coronavirus disease 2019. These treatments included antivirals, immune modulators, severe acute respiratory syndrome coronavirus 2 neutralizing antibodies, and organ-supportive treatments at both the preclinical and clinical stages of development. The ACTIV Therapeutics-Clinical Working Group Agent Prioritization subgroup established a uniform data collection process required to perform an assessment of any agent type using review criteria that were identified and differentially weighted for each agent class. The ACTIV Therapeutics-Clinical Working Group evaluated over 750 therapeutic agents with potential application for coronavirus disease 2019 and prioritized promising candidates for testing within the master protocols conducted by ACTIV. In addition, promising agents among preclinical candidates were selected by ACTIV to be matched with laboratories that could assist in executing rigorous preclinical studies. Between April 14, 2020, and May 31, 2021, the Agent Prioritization subgroup advanced 20 agents into the Accelerating Coronavirus Disease 2019 Therapeutic Interventions and Vaccines master protocols and matched 25 agents with laboratories to assist with preclinical testing.


Asunto(s)
Anticuerpos/uso terapéutico , Antivirales/uso terapéutico , Vacunas contra la COVID-19/administración & dosificación , COVID-19/prevención & control , COVID-19/terapia , Desarrollo de Medicamentos/organización & administración , Descubrimiento de Drogas/organización & administración , Humanos , National Institutes of Health (U.S.) , Pandemias , Asociación entre el Sector Público-Privado , SARS-CoV-2 , Estados Unidos , Tratamiento Farmacológico de COVID-19
4.
Am J Physiol Lung Cell Mol Physiol ; 318(2): L429-L441, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31850803

RESUMEN

Insulin resistance and right ventricular (RV) dysfunction are associated with lipotoxicity in heritable forms of pulmonary arterial hypertension (PAH), commonly due to mutations in bone morphogenetic protein receptor type 2 (BMPR2). How BMPR2 dysfunction in cardiomyocytes alters glucose metabolism and the response of these cells to insulin are unknown. We hypothesized that BMPR2 mutation in cardiomyocytes alters glucose-supported mitochondrial respiration and impairs cellular responses to insulin, including glucose and lipid uptake. We performed metabolic assays, immunofluorescence and Western analysis, RNA profiling, and radioactive isotope uptake studies in H9c2 cardiomyocyte cell lines with and without patient-derived BMPR2 mutations (mutant cells), with and without insulin. Unlike control cells, BMPR2 mutant cardiomyocytes have reduced metabolic plasticity as indicated by reduced mitochondrial respiration with increased mitochondrial superoxide production. These mutant cells show enhanced baseline phosphorylation of insulin-signaling protein as indicated by increased Akt, AMPK, and acetyl-CoA carboxylase phosphorylation that may negatively influence fatty acid oxidation and enhance lipid uptake, and are insulin insensitive. Furthermore, mutant cells demonstrate an increase in milk fat globule-EGF factor-8 protein (MFGE8), which influences the insulin-signaling pathway by phosphorylating AktSer473 via phosphatidylinositol 3-kinase and mammalian target of rapamycin. In conclusion, BMPR2 mutant cardiomyocytes have reduced metabolic plasticity and fail to respond to glucose. These cells have enhanced baseline insulin-signaling pattern favoring insulin resistance with failure to augment this pattern in response to insulin. BMPR2 mutation possibly blunts glucose uptake and enhances lipid uptake in these cardiomyocytes. The MFGE8-driven signaling pathway may suggest a new mechanism underlying RV lipotoxicity in PAH.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Miocitos Cardíacos/metabolismo , Transducción de Señal , Animales , Antígenos de Superficie/metabolismo , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Antígenos CD36/metabolismo , Línea Celular , Regulación de la Expresión Génica , Resistencia a la Insulina , Ratones , Proteínas de la Leche/metabolismo , Mitocondrias/metabolismo , Mutación/genética , Consumo de Oxígeno , Ácido Palmítico/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Ratas , Superóxidos/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
5.
Nucleic Acids Res ; 46(7): 3458-3467, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29438559

RESUMEN

Reactive oxygen species (ROS) are formed in mitochondria during electron transport and energy generation. Elevated levels of ROS lead to increased amounts of mitochondrial DNA (mtDNA) damage. We report that levels of M1dG, a major endogenous peroxidation-derived DNA adduct, are 50-100-fold higher in mtDNA than in nuclear DNA in several different human cell lines. Treatment of cells with agents that either increase or decrease mitochondrial superoxide levels leads to increased or decreased levels of M1dG in mtDNA, respectively. Sequence analysis of adducted mtDNA suggests that M1dG residues are randomly distributed throughout the mitochondrial genome. Basal levels of M1dG in mtDNA from pulmonary microvascular endothelial cells (PMVECs) from transgenic bone morphogenetic protein receptor 2 mutant mice (BMPR2R899X) (four adducts per 106 dG) are twice as high as adduct levels in wild-type cells. A similar increase was observed in mtDNA from heterozygous null (BMPR2+/-) compared to wild-type PMVECs. Pulmonary arterial hypertension is observed in the presence of BMPR2 signaling disruptions, which are also associated with mitochondrial dysfunction and oxidant injury to endothelial tissue. Persistence of M1dG adducts in mtDNA could have implications for mutagenesis and mitochondrial gene expression, thereby contributing to the role of mitochondrial dysfunction in diseases.


Asunto(s)
ADN Mitocondrial/metabolismo , Mitocondrias/genética , Estrés Oxidativo/genética , Nucleósidos de Purina/metabolismo , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Aductos de ADN/genética , Aductos de ADN/metabolismo , ADN Mitocondrial/genética , Transporte de Electrón/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Peroxidación de Lípido/genética , Ratones , Ratones Transgénicos , Mitocondrias/patología , Mutagénesis/genética , Oxidantes/farmacología , Nucleósidos de Purina/biosíntesis , Especies Reactivas de Oxígeno/química , Superóxidos/metabolismo
6.
Crit Care Med ; 47(8): 1065-1071, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31306254

RESUMEN

OBJECTIVES: Studies suggest that mitochondrial dysfunction underlies some forms of sepsis-induced organ failure. We sought to test the hypothesis that variations in mitochondrial DNA haplogroup affect susceptibility to sepsis-associated delirium, a common manifestation of acute brain dysfunction during sepsis. DESIGN: Retrospective cohort study. SETTING: Medical and surgical ICUs at a large tertiary care center. PATIENTS: Caucasian and African American adults with sepsis. MEASUREMENTS AND MAIN RESULTS: We determined each patient's mitochondrial DNA haplogroup using single-nucleotide polymorphisms genotyping data in a DNA databank and extracted outcomes from linked electronic medical records. We then used zero-inflated negative binomial regression to analyze age-adjusted associations between mitochondrial DNA haplogroups and duration of delirium, identified using the Confusion Assessment Method for the ICU. Eight-hundred ten patients accounted for 958 sepsis admissions, with 802 (84%) by Caucasians and 156 (16%) by African Americans. In total, 795 patient admissions (83%) involved one or more days of delirium. The 7% of Caucasians belonging to mitochondrial DNA haplogroup clade IWX experienced more delirium than the 49% in haplogroup H, the most common Caucasian haplogroup (age-adjusted rate ratio for delirium 1.36; 95% CI, 1.13-1.64; p = 0.001). Alternatively, among African Americans the 24% in haplogroup L2 experienced less delirium than those in haplogroup L3, the most common African haplogroup (adjusted rate ratio for delirium 0.60; 95% CI, 0.38-0.94; p = 0.03). CONCLUSIONS: Variations in mitochondrial DNA are associated with development of and protection from delirium in Caucasians and African Americans during sepsis. Future studies are now required to determine whether mitochondrial DNA and mitochondrial dysfunction contribute to the pathogenesis of delirium during sepsis so that targeted treatments can be developed.


Asunto(s)
Negro o Afroamericano/genética , ADN Mitocondrial/genética , Haplotipos/genética , Encefalopatía Asociada a la Sepsis/genética , Población Blanca/genética , Adulto , Enfermedad Crítica , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Estudios Retrospectivos , Análisis de Secuencia de ADN
7.
Circ Res ; 121(5): 564-574, 2017 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-28684630

RESUMEN

RATIONALE: Clinical studies have shown that Sirt3 (Sirtuin 3) expression declines by 40% by 65 years of age paralleling the increased incidence of hypertension and metabolic conditions further inactivate Sirt3 because of increased NADH (nicotinamide adenine dinucleotide, reduced form) and acetyl-CoA levels. Sirt3 impairment reduces the activity of a key mitochondrial antioxidant enzyme, superoxide dismutase 2 (SOD2) because of hyperacetylation. OBJECTIVE: In this study, we examined whether the loss of Sirt3 activity increases vascular oxidative stress because of SOD2 hyperacetylation and promotes endothelial dysfunction and hypertension. METHODS AND RESULTS: Hypertension was markedly increased in Sirt3-knockout (Sirt3-/-) and SOD2-depleted (SOD2+/-) mice in response to low dose of angiotensin II (0.3 mg/kg per day) compared with wild-type C57Bl/6J mice. Sirt3 depletion increased SOD2 acetylation, elevated mitochondrial O2· -, and diminished endothelial nitric oxide. Angiotensin II-induced hypertension was associated with Sirt3 S-glutathionylation, acetylation of vascular SOD2, and reduced SOD2 activity. Scavenging of mitochondrial H2O2 in mCAT mice expressing mitochondria-targeted catalase prevented Sirt3 and SOD2 impairment and attenuated hypertension. Treatment of mice after onset of hypertension with a mitochondria-targeted H2O2 scavenger, mitochondria-targeted hydrogen peroxide scavenger ebselen, reduced Sirt3 S-glutathionylation, diminished SOD2 acetylation, and reduced blood pressure in wild-type but not in Sirt3-/- mice, whereas an SOD2 mimetic, (2-[2,2,6,6-tetramethylpiperidin-1-oxyl-4-ylamino]-2-oxoethyl) triphenylphosphonium (mitoTEMPO), reduced blood pressure and improved vasorelaxation both in Sirt3-/- and wild-type mice. SOD2 acetylation had an inverse correlation with SOD2 activity and a direct correlation with the severity of hypertension. Analysis of human subjects with essential hypertension showed 2.6-fold increase in SOD2 acetylation and 1.4-fold decrease in Sirt3 levels, whereas SOD2 expression was not affected. CONCLUSIONS: Our data suggest that diminished Sirt3 expression and redox inactivation of Sirt3 lead to SOD2 inactivation and contributes to the pathogenesis of hypertension.


Asunto(s)
Hipertensión/metabolismo , Estrés Oxidativo/fisiología , Sirtuina 3/metabolismo , Superóxido Dismutasa/metabolismo , Acetilación , Animales , Células Cultivadas , Humanos , Hipertensión/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Sirtuina 3/genética , Superóxido Dismutasa/genética
8.
Biochem Biophys Res Commun ; 495(1): 433-437, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-29129689

RESUMEN

BACKGROUND: Increased endothelial permeability is central to shock and organ dysfunction in sepsis but therapeutics targeted to known mediators of increased endothelial permeability have been unsuccessful in patient studies. We previously reported that cell-free hemoglobin (CFH) is elevated in the majority of patients with sepsis and is associated with organ dysfunction, poor clinical outcomes and elevated markers of oxidant injury. Others have shown that Vitamin C (ascorbate) may have endothelial protective effects in sepsis. In this study, we tested the hypothesis that high levels of CFH, as seen in the circulation of patients with sepsis, disrupt endothelial barrier integrity. METHODS: Human umbilical vein endothelial cells (HUVEC) were grown to confluence and treated with CFH with or without ascorbate. Monolayer permeability was measured by Electric Cell-substrate Impedance Sensing (ECIS) or transfer of 14C-inulin. Viability was measured by trypan blue exclusion. Intracellular ascorbate was measured by HPLC. RESULTS: CFH increased permeability in a dose- and time-dependent manner with 1 mg/ml of CFH increasing inulin transfer by 50% without affecting cell viability. CFH (1 mg/ml) also caused a dramatic reduction in intracellular ascorbate in the same time frame (1.4 mM without CFH, 0.23 mM 18 h after 1 mg/ml CFH, p < 0.05). Pre-treatment of HUVECs with ascorbate attenuated CFH induced permeability. CONCLUSIONS: CFH increases endothelial permeability in part through depletion of intracellular ascorbate. Supplementation of ascorbate can attenuate increases in permeability mediated by CFH suggesting a possible therapeutic approach in sepsis.


Asunto(s)
Antioxidantes/farmacología , Ácido Ascórbico/farmacología , Permeabilidad Capilar/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Hemoglobinas/metabolismo , Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Sepsis/tratamiento farmacológico , Sepsis/metabolismo
9.
Eur Respir J ; 51(6)2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29903860

RESUMEN

Pulmonary arterial hypertension (PAH) is a deadly disease with no cure. Alternate conversion of angiotensin II (AngII) to angiotensin-(1-7) (Ang-(1-7)) by angiotensin-converting enzyme 2 (ACE2) resulting in Mas receptor (Mas1) activation improves rodent models of PAH. Effects of recombinant human (rh) ACE2 in human PAH are unknown. Our objective was to determine the effects of rhACE2 in PAH.We defined the molecular effects of Mas1 activation using porcine pulmonary arteries, measured AngII/Ang-(1-7) levels in human PAH and conducted a phase IIa, open-label pilot study of a single infusion of rhACE2 (GSK2586881, 0.2 or 0.4 mg·kg-1 intravenously).Superoxide dismutase 2 (SOD2) and inflammatory gene expression were identified as markers of Mas1 activation. After confirming reduced plasma ACE2 activity in human PAH, five patients were enrolled in the trial. GSK2586881 was well tolerated with significant improvement in cardiac output and pulmonary vascular resistance. GSK2586881 infusion was associated with reduced plasma markers of inflammation within 2-4 h and increased SOD2 plasma protein at 2 weeks.PAH is characterised by reduced ACE2 activity. Augmentation of ACE2 in a pilot study was well tolerated, associated with improved pulmonary haemodynamics and reduced markers of oxidant and inflammatory mediators. Targeting this pathway may be beneficial in human PAH.


Asunto(s)
Citocinas/metabolismo , Hipertensión Pulmonar/tratamiento farmacológico , Peptidil-Dipeptidasa A/farmacología , Arteria Pulmonar/fisiopatología , Adulto , Anciano , Enzima Convertidora de Angiotensina 2 , Animales , Biomarcadores , Citocinas/efectos de los fármacos , Femenino , Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Proyectos Piloto , Prueba de Estudio Conceptual , Proto-Oncogenes Mas , Proteínas Proto-Oncogénicas/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Superóxido Dismutasa/metabolismo , Porcinos , Resistencia Vascular/efectos de los fármacos
10.
Circulation ; 133(20): 1936-44, 2016 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-27006481

RESUMEN

BACKGROUND: The mechanisms of right ventricular (RV) failure in pulmonary arterial hypertension (PAH) are poorly understood. Abnormalities in fatty acid (FA) metabolism have been described in experimental models of PAH, but systemic and myocardial FA metabolism has not been studied in human PAH. METHODS AND RESULTS: We used human blood, RV tissue, and noninvasive imaging to characterize multiple steps in the FA metabolic pathway in PAH subjects and controls. Circulating free FAs and long-chain acylcarnitines were elevated in PAH patients versus controls. Human RV long-chain FAs were increased and long-chain acylcarnitines were markedly reduced in PAH versus controls. With the use of proton magnetic resonance spectroscopy, in vivo myocardial triglyceride content was elevated in human PAH versus controls (1.4±1.3% triglyceride versus 0.22±0.11% triglyceride, P=0.02). Ceramide, a mediator of lipotoxicity, was increased in PAH RVs versus controls. Using an animal model of heritable PAH, we demonstrated reduced FA oxidation via failure of palmitoylcarnitine to stimulate oxygen consumption in the PAH RV. CONCLUSIONS: Abnormalities in FA metabolism can be detected in the blood and myocardium in human PAH and are associated with in vivo cardiac steatosis and lipotoxicity. Murine data suggest that lipotoxicity may arise from reduction in FA oxidation.


Asunto(s)
Ácidos Grasos/metabolismo , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Triglicéridos/metabolismo , Disfunción Ventricular Derecha/metabolismo , Disfunción Ventricular Derecha/patología , Animales , Ceramidas/metabolismo , Estudios de Cohortes , Humanos , Hipertensión Pulmonar/epidemiología , Ratones , Ratones Transgénicos , Estudios Prospectivos , Disfunción Ventricular Derecha/epidemiología
11.
Circulation ; 133(1): 82-97, 2016 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-26487756

RESUMEN

BACKGROUND: Pulmonary arterial hypertension (PAH) is a proliferative disease of the pulmonary vasculature that preferentially affects women. Estrogens such as the metabolite 16α-hydroxyestrone (16αOHE) may contribute to PAH pathogenesis, and alterations in cellular energy metabolism associate with PAH. We hypothesized that 16αOHE promotes heritable PAH (HPAH) via microRNA-29 (miR-29) family upregulation and that antagonism of miR-29 would attenuate pulmonary hypertension in transgenic mouse models of Bmpr2 mutation. METHODS AND RESULTS: MicroRNA array profiling of human lung tissue found elevation of microRNAs associated with energy metabolism, including the miR-29 family, among HPAH patients. miR-29 expression was 2-fold higher in Bmpr2 mutant mice lungs at baseline compared with controls and 4 to 8-fold higher in Bmpr2 mice exposed to 16αOHE 1.25 µg/h for 4 weeks. Blot analyses of Bmpr2 mouse lung protein showed significant reductions in peroxisome proliferator-activated receptor-γ and CD36 in those mice exposed to 16αOHE and protein derived from HPAH lungs compared with controls. Bmpr2 mice treated with anti-miR-29 (20-mg/kg injections for 6 weeks) had improvements in hemodynamic profile, histology, and markers of dysregulated energy metabolism compared with controls. Pulmonary artery smooth muscle cells derived from Bmpr2 murine lungs demonstrated mitochondrial abnormalities, which improved with anti-miR-29 transfection in vitro; endothelial-like cells derived from HPAH patient induced pluripotent stem cell lines were similar and improved with anti-miR-29 treatment. CONCLUSIONS: 16αOHE promotes the development of HPAH via upregulation of miR-29, which alters molecular and functional indexes of energy metabolism. Antagonism of miR-29 improves in vivo and in vitro features of HPAH and reveals a possible novel therapeutic target.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/biosíntesis , Microambiente Celular/fisiología , Hidroxiestronas/metabolismo , Hipertensión Pulmonar/metabolismo , MicroARNs/biosíntesis , Animales , Microambiente Celular/efectos de los fármacos , Femenino , Humanos , Hidroxiestronas/toxicidad , Hipertensión Pulmonar/inducido químicamente , Hipertensión Pulmonar/diagnóstico , Masculino , Ratones , Ratones Transgénicos , MicroARNs/antagonistas & inhibidores
12.
Pharmacogenet Genomics ; 27(7): 247-254, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28542097

RESUMEN

BACKGROUND: Anthracyclines are important chemotherapeutic agents, but their use is limited by cardiotoxicity. Candidate gene and genome-wide studies have identified putative risk loci for overt cardiotoxicity and heart failure, but there has been no comprehensive assessment of genomic variation influencing the intermediate phenotype of anthracycline-related changes in left ventricular (LV) function. The purpose of this study was to identify genetic factors influencing changes in LV function after anthracycline chemotherapy. METHODS: We conducted a genome-wide association study (GWAS) of change in LV function after anthracycline exposure in 385 patients identified from BioVU, a resource linking DNA samples to de-identified electronic medical record data. Variants with P values less than 1×10 were independently tested for replication in a cohort of 181 anthracycline-exposed patients from a prospective clinical trial. Pathway analysis was performed to assess combined effects of multiple genetic variants. RESULTS: Both cohorts were middle-aged adults of predominantly European descent. Among 11 candidate loci identified in discovery GWAS, one single nucleotide polymorphism near PR domain containing 2, with ZNF domain (PRDM2), rs7542939, had a combined P value of 6.5×10 in meta-analysis. Eighteen Kyoto Encyclopedia of Gene and Genomes pathways showed strong enrichment for variants associated with the primary outcome. Identified pathways related to DNA repair, cellular metabolism, and cardiac remodeling. CONCLUSION: Using genome-wide association we identified a novel candidate susceptibility locus near PRDM2. Variation in genes belonging to pathways related to DNA repair, metabolism, and cardiac remodeling may influence changes in LV function after anthracycline exposure.


Asunto(s)
Antraciclinas/farmacología , Estudio de Asociación del Genoma Completo , Transducción de Señal/genética , Función Ventricular Izquierda/efectos de los fármacos , Función Ventricular Izquierda/genética , Adulto , Estudios de Cohortes , Demografía , Femenino , Humanos , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Volumen Sistólico/genética
13.
Eur Respir J ; 50(2)2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28775043

RESUMEN

Increased oestrogen is a strong epidemiological risk factor for development of pulmonary arterial hypertension (PAH) in patients, associated with metabolic defects. In addition, oestrogens drive penetrance in mice carrying mutations in bone morphogenetic protein receptor type II (BMPR2), the cause of most heritable PAH. The goal of the present study was to determine whether inhibition of oestrogens was effective in the treatment of PAH in these mice.The oestrogen inhibitors fulvestrant and anastrozole were used in a prevention and treatment paradigm in BMPR2 mutant mice, and tamoxifen was used for treatment. In addition, BMPR2 mutant mice were crossed onto oestrogen receptor (ESR)1 and ESR2 knockout backgrounds to assess receptor specificity. Haemodynamic and metabolic outcomes were measured.Oestrogen inhibition both prevented and treated PAH in BMPR2 mutant mice. This was associated with reduction in metabolic defects including oxidised lipid formation, insulin resistance and rescue of peroxisome proliferator-activated receptor-γ and CD36. The effect was mediated primarily through ESR2, but partially through ESR1.Our data suggest that trials of oestrogen inhibition in human PAH are warranted, and may improve pulmonary vascular disease through amelioration of metabolic defects. Although fulvestrant and anastrozole were more effective than tamoxifen, tamoxifen may be useful in premenopausal females, because of a reduced risk of induction of menopause.


Asunto(s)
Antagonistas de Estrógenos/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/fisiopatología , Tamoxifeno/farmacología , Anastrozol , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Modelos Animales de Enfermedad , Ecocardiografía , Estradiol/análogos & derivados , Estradiol/sangre , Estradiol/farmacología , Femenino , Fulvestrant , Hemodinámica , Humanos , Resistencia a la Insulina , Pulmón/patología , Ratones , Ratones Noqueados , Mutación , Nitrilos/farmacología , Transducción de Señal/efectos de los fármacos , Triazoles/farmacología
14.
Am J Respir Crit Care Med ; 194(6): 719-28, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27077479

RESUMEN

RATIONALE: In heritable pulmonary arterial hypertension with germline mutation in the bone morphogenetic protein receptor type 2 (BMPR2) gene, right ventricle (RV) dysfunction is associated with RV lipotoxicity; however, the underlying mechanism for lipid accumulation is not known. OBJECTIVES: We hypothesized that lipid accumulation in cardiomyocytes with BMPR2 mutation occurs owing to alterations in lipid transport and impaired fatty acid oxidation (FAO), which is exacerbated by a high-lipid (Western) diet (WD). METHODS: We used a transgenic mouse model of pulmonary arterial hypertension with mutant BMPR2 and generated a cardiomyocyte cell line with BMPR2 mutation. Electron microscopy and metabolomic analysis were performed on mouse RVs. MEASUREMENTS AND MAIN RESULTS: By metabolomics analysis, we found an increase in long-chain fatty acids in BMPR2 mutant mouse RVs compared with controls, which correlated with cardiac index. BMPR2-mutant cardiomyocytes had increased lipid compared with controls. Direct measurement of FAO in the WD-fed BMPR2-mutant RV showed impaired palmitate-linked oxygen consumption, and metabolomics analysis showed reduced indices of FAO. Using both mutant BMPR2 mouse RVs and cardiomyocytes, we found an increase in the uptake of (14)C-palmitate and fatty acid transporter CD36 that was further exacerbated by WD. CONCLUSIONS: Taken together, our data suggest that impaired FAO and increased expression of the lipid transporter CD36 are key mechanisms underlying lipid deposition in the BMPR2-mutant RV, which are exacerbated in the presence of dietary lipids. These findings suggest important features leading to RV lipotoxicity in pulmonary arterial hypertension and may point to novel areas of therapeutic intervention.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Ventrículos Cardíacos/química , Lípidos/análisis , Animales , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/fisiología , Línea Celular , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Ventrículos Cardíacos/metabolismo , Ventrículos Cardíacos/ultraestructura , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Metabolismo de los Lípidos/genética , Metabolómica , Ratones , Ratones Transgénicos , Microscopía Electrónica , Miocitos Cardíacos/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 310(3): L249-62, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26637636

RESUMEN

Pulmonary hypertension (PH) complicating chronic parenchymal lung disease, such as idiopathic pulmonary fibrosis, results in significant morbidity and mortality. Since the hypoxia-inducible factor (HIF) signaling pathway is important for development of pulmonary hypertension in chronic hypoxia, we investigated whether HIF signaling in vascular endothelium regulates development of PH related to pulmonary fibrosis. We generated a transgenic model in which HIF is deleted within vascular endothelial cells and then exposed these mice to chronic intraperitoneal bleomycin to induce PH associated with lung fibrosis. Although no differences in the degree of fibrotic remodeling were observed, we found that endothelial HIF-deficient mice were protected against development of PH, including right ventricle and pulmonary vessel remodeling. Similarly, endothelial HIF-deficient mice were protected from PH after a 4-wk exposure to normobaric hypoxia. In vitro studies of pulmonary vascular endothelial cells isolated from the HIF-targeted mice and controls revealed that endothelial HIF signaling increases endothelial cell expression of connective tissue growth factor, enhances vascular permeability, and promotes pulmonary artery smooth muscle cell proliferation and wound healing ability, all of which have the potential to impact the development of PH in vivo. Taken together, these studies demonstrate that vascular endothelial cell HIF signaling is necessary for development of hypoxia and pulmonary fibrosis associated PH. As such, HIF and HIF-regulated targets represent a therapeutic target in these conditions.


Asunto(s)
Células Endoteliales/metabolismo , Hipertensión Pulmonar/metabolismo , Factor 1 Inducible por Hipoxia/metabolismo , Arteria Pulmonar/metabolismo , Animales , Proliferación Celular/fisiología , Células Cultivadas , Endotelio Vascular/metabolismo , Fibrosis/etiología , Hipertensión Pulmonar/complicaciones , Hipoxia/metabolismo , Ratones Transgénicos , Músculo Liso Vascular/metabolismo , Remodelación Vascular/fisiología
16.
Am J Respir Crit Care Med ; 189(3): 325-34, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24274756

RESUMEN

RATIONALE: Shorter survival in heritable pulmonary arterial hypertension (HPAH), often due to BMPR2 mutation, has been described in association with impaired right ventricle (RV) compensation. HPAH animal models are insulin resistant, and cells with BMPR2 mutation have impaired fatty acid oxidation, but whether these findings affect the RV in HPAH is unknown. OBJECTIVES: To test the hypothesis that BMPR2 mutation impairs RV hypertrophic responses in association with lipid deposition. METHODS: RV hypertrophy was assessed in two models of mutant Bmpr2 expression, smooth muscle-specific (Sm22(R899X)) and universal expression (Rosa26(R899X)). Littermate control mice underwent the same stress using pulmonary artery banding (Low-PAB). Lipid content was assessed in rodent and human HPAH RVs and in Rosa26(R899X) mice after metformin administration. RV microarrays were performed using human HPAH and control subjects. RESULTS: RV/(left ventricle + septum) did not rise directly in proportion to RV systolic pressure in Rosa26(R899X) but did in Sm22(R899X) (P < 0.05). Rosa26(R899X) RVs demonstrated intracardiomyocyte triglyceride deposition not present in Low-PAB (P < 0.05). RV lipid deposition was identified in human HPAH RVs but not in controls. Microarray analysis demonstrated defects in fatty acid oxidation in human HPAH RVs. Metformin in Rosa26(R899X) mice resulted in reduced RV lipid deposition. CONCLUSIONS: These data demonstrate that Bmpr2 mutation affects RV stress responses in a transgenic rodent model. Impaired RV hypertrophy and triglyceride and ceramide deposition are present as a function of RV mutant Bmpr2 in mice; fatty acid oxidation impairment in human HPAH RVs may underlie this finding. Further study of how BMPR2 mediates RV lipotoxicity is warranted.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Ceramidas/metabolismo , Hipertensión Pulmonar/genética , Hipertrofia Ventricular Derecha/metabolismo , Lipólisis , Triglicéridos/metabolismo , Animales , Biomarcadores/metabolismo , Estudios de Casos y Controles , Hipertensión Pulmonar Primaria Familiar , Marcadores Genéticos , Humanos , Hipertensión Pulmonar/complicaciones , Hipertrofia Ventricular Derecha/etiología , Hipertrofia Ventricular Derecha/genética , Ratones , Ratones Transgénicos , Mutación , Análisis de Secuencia por Matrices de Oligonucleótidos , Oxidación-Reducción
17.
Am J Physiol Cell Physiol ; 307(5): C415-30, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24871858

RESUMEN

Understanding differences in gene expression that increase risk for pulmonary arterial hypertension (PAH) is essential to understanding the molecular basis for disease. Previous studies on patient samples were limited by end-stage disease effects or by use of nonadherent cells, which are not ideal to model vascular cells in vivo. These studies addressed the hypothesis that pathological processes associated with PAH may be identified via a genetic signature common across multiple cell types. Expression array experiments were initially conducted to analyze cell types at different stages of vascular differentiation (mesenchymal stromal and endothelial) derived from PAH patient-specific induced pluripotent stem (iPS) cells. Molecular pathways that were altered in the PAH cell lines were then compared with those in fibroblasts from 21 patients, including those with idiopathic and heritable PAH. Wnt was identified as a target pathway and was validated in vitro using primary patient mesenchymal and endothelial cells. Taken together, our data suggest that the molecular lesions that cause PAH are present in all cell types evaluated, regardless of origin, and that stimulation of the Wnt signaling pathway was a common molecular defect in both heritable and idiopathic PAH.


Asunto(s)
Diferenciación Celular/genética , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/patología , Células Madre Pluripotentes/patología , Vía de Señalización Wnt/genética , Línea Celular , Células Cultivadas , Células Endoteliales/patología , Células Endoteliales/fisiología , Hipertensión Pulmonar Primaria Familiar , Humanos , Células Madre Pluripotentes/fisiología , Mucosa Respiratoria/patología , Mucosa Respiratoria/fisiología
19.
Am J Respir Cell Mol Biol ; 49(5): 778-87, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23742019

RESUMEN

Pulmonary arterial hypertension (PAH) has been associated with a number of different but interrelated pathogenic mechanisms. Metabolic and oxidative stresses have been shown to play important pathogenic roles in a variety of model systems. However, many of these relationships remain at the level of association. We sought to establish a direct role for metabolic stress and oxidant injury in the pathogenesis of PAH. Mice that universally express a disease-causing mutation in bone morphogenic protein receptor 2 (Bmpr2) were exposed to room air or to brief daily hyperoxia (95% oxygen for 3 h) for 6 weeks, and were compared with wild-type animals undergoing identical exposures. In both murine tissues and cultured endothelial cells, the expression of mutant Bmpr2 was sufficient to cause oxidant injury that was particularly pronounced in mitochondrial membranes. With the enhancement of mitochondrial generation of reactive oxygen species by hyperoxia, oxidant injury was substantially enhanced in mitochondrial membranes, even in tissues distant from the lung. Hyperoxia, despite its vasodilatory actions in the pulmonary circulation, significantly worsened the PAH phenotype (elevated right ventricular systolic pressure, decreased cardiac output, and increased pulmonary vascular occlusion) in Bmpr2 mutant animals. These experiments demonstrate that oxidant injury and metabolic stress contribute directly to disease development, and provide further evidence for PAH as a systemic disease with life-limiting cardiopulmonary manifestations.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/metabolismo , Células Endoteliales/metabolismo , Hiperoxia/complicaciones , Hipertensión Pulmonar/etiología , Lesión Pulmonar/etiología , Pulmón/irrigación sanguínea , Mutación , Estrés Oxidativo , Estrés Fisiológico , Animales , Presión Arterial , Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Gasto Cardíaco , Línea Celular Tumoral , Modelos Animales de Enfermedad , Células Endoteliales/patología , Hipertensión Pulmonar Primaria Familiar , Humanos , Hiperoxia/genética , Hiperoxia/metabolismo , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Hipertensión Pulmonar/fisiopatología , Lesión Pulmonar/genética , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Lesión Pulmonar/fisiopatología , Ratones , Ratones Transgénicos , Mitocondrias/metabolismo , Mitocondrias/patología , Membranas Mitocondriales/metabolismo , Membranas Mitocondriales/patología , Arteria Pulmonar/metabolismo , Arteria Pulmonar/fisiopatología , Especies Reactivas de Oxígeno/metabolismo , Función Ventricular Derecha , Presión Ventricular
20.
Eur Respir J ; 41(4): 861-71, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22936709

RESUMEN

Patients with pulmonary arterial hypertension have increased prevalence of insulin resistance. We aimed to determine whether metabolic defects are associated with bone morphogenic protein receptor type 2 (Bmpr2) mutations in mice, and whether these may contribute to pulmonary vascular disease development. Metabolic phenotyping was performed on transgenic mice with inducible expression of Bmpr2 mutation, R899X. Phenotypic penetrance in Bmpr2(R899X) was assessed in a high-fat diet model of insulin resistance. Alterations in glucocorticoid responses were assessed in murine pulmonary microvascular endothelial cells and Bmpr2(R899X) mice treated with dexamethasone. Compared to controls, Bmpr2(R899X) mice showed increased weight gain and demonstrated insulin resistance as assessed by the homeostatic model assessment insulin resistance (1.0 ± 0.4 versus 2.2 ± 1.8) and by fat accumulation in skeletal muscle and decreased oxygen consumption. Bmpr2(R899X) mice fed a high-fat diet had strong increases in pulmonary hypertension penetrance (seven out of 11 versus three out of 11). In cell culture and in vivo experiments, Bmpr2 mutation resulted in a combination of constitutive glucocorticoid receptor activation and insensitivity. Insulin resistance is present as an early feature of Bmpr2 mutation in mice. Exacerbated insulin resistance through high-fat diet worsened pulmonary phenotype, implying a possible causal role in disease. Impaired glucocorticoid responses may contribute to metabolic defects.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo II/genética , Hipertensión Pulmonar/tratamiento farmacológico , Resistencia a la Insulina , Animales , Composición Corporal , Línea Celular , Dexametasona/farmacología , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Metabolismo Energético , Humanos , Hipertensión Pulmonar/metabolismo , Masculino , Ratones , Ratones Transgénicos , Mutación , Fenotipo , Transporte de Proteínas , Ratas , Receptores de Glucocorticoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA