Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 110
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 84(7): 1321-1337.e11, 2024 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-38513662

RESUMEN

Intracellular Mg2+ (iMg2+) is bound with phosphometabolites, nucleic acids, and proteins in eukaryotes. Little is known about the intracellular compartmentalization and molecular details of Mg2+ transport into/from cellular organelles such as the endoplasmic reticulum (ER). We found that the ER is a major iMg2+ compartment refilled by a largely uncharacterized ER-localized protein, TMEM94. Conventional and AlphaFold2 predictions suggest that ERMA (TMEM94) is a multi-pass transmembrane protein with large cytosolic headpiece actuator, nucleotide, and phosphorylation domains, analogous to P-type ATPases. However, ERMA uniquely combines a P-type ATPase domain and a GMN motif for ERMg2+ uptake. Experiments reveal that a tyrosine residue is crucial for Mg2+ binding and activity in a mechanism conserved in both prokaryotic (mgtB and mgtA) and eukaryotic Mg2+ ATPases. Cardiac dysfunction by haploinsufficiency, abnormal Ca2+ cycling in mouse Erma+/- cardiomyocytes, and ERMA mRNA silencing in human iPSC-cardiomyocytes collectively define ERMA as an essential component of ERMg2+ uptake in eukaryotes.


Asunto(s)
Adenosina Trifosfatasas , ATPasas Tipo P , Animales , Ratones , Humanos , Adenosina Trifosfatasas/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Retículo Endoplásmico/genética , Retículo Endoplásmico/metabolismo , Transporte Biológico , ATPasas Tipo P/metabolismo , Calcio/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico
2.
Cell ; 154(3): 556-68, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23911321

RESUMEN

Skp2 E3 ligase is overexpressed in numerous human cancers and plays a critical role in cell-cycle progression, senescence, metabolism, cancer progression, and metastasis. In the present study, we identified a specific Skp2 inhibitor using high-throughput in silico screening of large and diverse chemical libraries. This Skp2 inhibitor selectively suppresses Skp2 E3 ligase activity, but not activity of other SCF complexes. It also phenocopies the effects observed upon genetic Skp2 deficiency, such as suppressing survival and Akt-mediated glycolysis and triggering p53-independent cellular senescence. Strikingly, we discovered a critical function of Skp2 in positively regulating cancer stem cell populations and self-renewal ability through genetic and pharmacological approaches. Notably, Skp2 inhibitor exhibits potent antitumor activities in multiple animal models and cooperates with chemotherapeutic agents to reduce cancer cell survival. Our study thus provides pharmacological evidence that Skp2 is a promising target for restricting cancer stem cell and cancer progression.


Asunto(s)
Antineoplásicos/farmacología , Descubrimiento de Drogas , Neoplasias/enzimología , Células Madre Neoplásicas/efectos de los fármacos , Proteínas Quinasas Asociadas a Fase-S/antagonistas & inhibidores , Ubiquitina-Proteína Ligasas/antagonistas & inhibidores , Animales , Antineoplásicos/química , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Genes p53 , Glucólisis/efectos de los fármacos , Humanos , Ratones , Ratones Desnudos , Modelos Moleculares , Complejos Multienzimáticos/antagonistas & inhibidores , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , Trasplante de Neoplasias , Neoplasias/tratamiento farmacológico , Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Proteínas Quinasas Asociadas a Fase-S/química , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Trasplante Heterólogo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo
3.
Angew Chem Int Ed Engl ; 62(37): e202308004, 2023 09 11.
Artículo en Inglés | MEDLINE | ID: mdl-37455289

RESUMEN

Small-molecule stabilization of protein-protein interactions (PPIs) is a promising strategy in chemical biology and drug discovery. However, the systematic discovery of PPI stabilizers remains a largely unmet challenge. Herein we report a fragment-linking approach targeting the interface of 14-3-3 and a peptide derived from the estrogen receptor alpha (ERα) protein. Two classes of fragments-a covalent and a noncovalent fragment-were co-crystallized and subsequently linked, resulting in a noncovalent hybrid molecule in which the original fragment interactions were largely conserved. Supported by 20 crystal structures, this initial hybrid molecule was further optimized, resulting in selective, 25-fold stabilization of the 14-3-3/ERα interaction. The high-resolution structures of both the single fragments, their co-crystal structures and those of the linked fragments document a feasible strategy to develop orthosteric PPI stabilizers by linking to an initial tethered fragment.


Asunto(s)
Proteínas 14-3-3 , Receptor alfa de Estrógeno , Proteínas 14-3-3/química , Receptor alfa de Estrógeno/metabolismo , Unión Proteica , Descubrimiento de Drogas/métodos
4.
Hum Mol Genet ; 28(15): 2486-2500, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31009944

RESUMEN

Mutations in LMNA encoding lamin A/C and EMD encoding emerin cause cardiomyopathy and muscular dystrophy. Lmna null mice develop these disorders and have a lifespan of 7-8 weeks. Emd null mice show no overt pathology and have normal skeletal muscle but with regeneration defects. We generated mice with germline deletions of both Lmna and Emd to determine the effects of combined loss of the encoded proteins. Mice without lamin A/C and emerin are born at the expected Mendelian ratio, are grossly normal at birth but have shorter lifespans than those lacking only lamin A/C. However, there are no major differences between these mice with regards to left ventricular function, heart ultrastructure or electrocardiographic parameters except for slower heart rates in the mice lacking both lamin A/C and emerin. Skeletal muscle is similarly affected in both of these mice. Lmna+/- mice also lacking emerin live to at least 1 year and have no significant differences in growth, heart or skeletal muscle compared to Lmna+/- mice. Deletion of the mouse gene encoding lamina-associated protein 1 leads to prenatal death; however, mice with heterozygous deletion of this gene lacking both lamin A/C and emerin are born at the expected Mendelian ratio but had a shorter lifespan than those only lacking lamin A/C and emerin. These results show that mice with combined deficiencies of three interacting nuclear envelope proteins have normal embryonic development and that early postnatal defects are primarily driven by loss of lamin A/C or lamina-associated polypeptide 1 rather than emerin.


Asunto(s)
Proteínas Portadoras/genética , Corazón/fisiopatología , Lamina Tipo A/genética , Proteínas de la Membrana/genética , Músculo Esquelético/fisiopatología , Distrofia Muscular de Emery-Dreifuss/genética , Mutación , Proteínas Nucleares/genética , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Femenino , Haploinsuficiencia , Masculino , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Distrofia Muscular de Emery-Dreifuss/fisiopatología , Miocardio/metabolismo , Miocardio/patología
5.
Am J Physiol Heart Circ Physiol ; 318(4): H778-H786, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32142354

RESUMEN

Sepsis-induced cardiomyopathy (SIC) is associated with increased patient mortality. At present, there are no specific therapies for SIC. Previous studies have reported increased reactive oxygen species (ROS) and mitochondrial dysfunction during SIC. However, a unifying mechanism remains to be defined. We hypothesized that PKCδ is required for abnormal calcium handling and cardiac mitochondrial dysfunction during sepsis and that genetic deletion of PKCδ would be protective. Polymicrobial sepsis induced by cecal ligation and puncture (CLP) surgery decreased the ejection fraction of wild-type (WT) mice but not PKCδ knockout (KO) mice. Similarly, WT cardiomyocytes exposed to lipopolysaccharide (LPS) demonstrated decreases in contractility and calcium transient amplitude that were not observed in PKCδ KO cardiomyocytes. LPS treatment decreased sarcoplasmic reticulum calcium stores in WT cardiomyocytes, which correlated with increased ryanodine receptor-2 oxidation in WT hearts but not PKCδ KO hearts after sepsis. LPS exposure increased mitochondrial ROS and decreased mitochondrial inner membrane potential in WT cardiomyocytes. This corresponded to morphologic changes consistent with mitochondrial dysfunction such as decreased overall size and cristae disorganization. Increased cellular ROS and changes in mitochondrial morphology were not observed in PKCδ KO cardiomyocytes. These data show that PKCδ is required in the pathophysiology of SIC by generating ROS and promoting mitochondrial dysfunction. Thus, PKCδ is a potential target for cardiac protection during sepsis.NEW & NOTEWORTHY Sepsis is often complicated by cardiac dysfunction, which is associated with a high mortality rate. Our work shows that the protein PKCδ is required for decreased cardiac contractility during sepsis. Mice with deletion of PKCδ are protected from cardiac dysfunction after sepsis. PKCδ causes mitochondrial dysfunction in cardiac myocytes, and reducing mitochondrial oxidative stress improves contractility in wild-type cardiomyocytes. Thus, PKCδ is a potential target for cardiac protection during sepsis.


Asunto(s)
Cardiomiopatías/genética , Mitocondrias Cardíacas/metabolismo , Proteína Quinasa C-delta/genética , Sepsis/complicaciones , Animales , Señalización del Calcio , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Células Cultivadas , Femenino , Eliminación de Gen , Lipopolisacáridos/toxicidad , Masculino , Potencial de la Membrana Mitocondrial , Ratones , Contracción Miocárdica , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Estrés Oxidativo , Proteína Quinasa C-delta/metabolismo
6.
Biochem Biophys Res Commun ; 527(4): 979-984, 2020 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-32439159

RESUMEN

Hepatic γ-secretase regulates low-density lipoprotein receptor (LDLR) cleavage and degradation, affecting clearance of plasma triglyceride (TG)-rich lipoproteins (TRLs). In this study, we investigated whether γ-secretase inhibition modulates risk of Western (high-fat/sucrose and high-cholesterol)-type diet (WTD)-induced hepatic steatosis, dyslipidemia and atherosclerosis. We evaluated liver and plasma lipids in WTD-fed mice with hepatocyte-specific ablation of the non-redundant γ-secretase-targeting subunit Nicastrin (L-Ncst). In parallel, we investigated the effect of liver-selective Ncst antisense oligonucleotides (ASO) on lipid metabolism and atherosclerosis in wildtype (WT) and ApoE knockout (ApoE-/-) mice fed normal chow or WTD. WTD-fed L-Ncst and Ncst ASO-treated WT mice showed reduced total cholesterol and LDL-cholesterol (LDL-C), as well as reduced hepatic lipid content as compared to Cre- and control ASO-treated WT mice. Treatment of WTD-fed ApoE-/- mice with Ncst ASO markedly lowered total and LDL cholesterol, hepatic TG and attenuated atherosclerotic lesions in the aorta, as compared to control ASO-treated mice. L-Ncst and Ncst ASO similarly showed reduced plasma glucose as compared to control mice. In conclusion, inhibition of hepatic γ-secretase reduces plasma glucose, and attenuates WTD-induced dyslipidemia, hepatic fat accumulation and atherosclerosis, suggesting potential pleiotropic application for diet-induced metabolic dysfunction.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/genética , Aterosclerosis/terapia , Dislipidemias/terapia , Hígado Graso/terapia , Glicoproteínas de Membrana/genética , Oligonucleótidos Antisentido/uso terapéutico , Animales , Aterosclerosis/sangre , Aterosclerosis/etiología , Aterosclerosis/genética , Dieta Occidental/efectos adversos , Dislipidemias/sangre , Dislipidemias/etiología , Dislipidemias/genética , Hígado Graso/sangre , Hígado Graso/etiología , Hígado Graso/genética , Técnicas de Inactivación de Genes , Terapia Genética , Lípidos/análisis , Lípidos/sangre , Hígado/metabolismo , Hígado/patología , Masculino , Ratones , Ratones Endogámicos C57BL
7.
J Cardiovasc Electrophysiol ; 31(12): 3086-3096, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33022765

RESUMEN

INTRODUCTION: Electrocardiographic characteristics in COVID-19-related mortality have not yet been reported, particularly in racial/ethnic minorities. METHODS AND RESULTS: We reviewed demographics, laboratory and cardiac tests, medications, and cardiac rhythm proximate to death or initiation of comfort care for patients hospitalized with a positive SARS-CoV-2 reverse-transcriptase polymerase chain reaction in three New York City hospitals between March 1 and April 3, 2020 who died. We described clinical characteristics and compared factors contributing toward arrhythmic versus nonarrhythmic death. Of 1258 patients screened, 133 died and were enrolled. Of these, 55.6% (74/133) were male, 69.9% (93/133) were racial/ethnic minorities, and 88.0% (117/133) had cardiovascular disease. The last cardiac rhythm recorded was VT or fibrillation in 5.3% (7/133), pulseless electrical activity in 7.5% (10/133), unspecified bradycardia in 0.8% (1/133), and asystole in 26.3% (35/133). Most 74.4% (99/133) died receiving comfort measures only. The most common abnormalities on admission electrocardiogram included abnormal QRS axis (25.8%), atrial fibrillation/flutter (14.3%), atrial ectopy (12.0%), and right bundle branch block (11.9%). During hospitalization, an additional 17.6% developed atrial ectopy, 14.7% ventricular ectopy, 10.1% atrial fibrillation/flutter, and 7.8% a right ventricular abnormality. Arrhythmic death was confirmed or suspected in 8.3% (11/133) associated with age, coronary artery disease, asthma, vasopressor use, longer admission corrected QT interval, and left bundle branch block (LBBB). CONCLUSIONS: Conduction, rhythm, and electrocardiographic abnormalities were common during COVID-19-related hospitalization. Arrhythmic death was associated with age, coronary artery disease, asthma, longer admission corrected QT interval, LBBB, ventricular ectopy, and usage of vasopressors. Most died receiving comfort measures.


Asunto(s)
Arritmias Cardíacas/mortalidad , COVID-19/mortalidad , Mortalidad Hospitalaria , Anciano , Anciano de 80 o más Años , Arritmias Cardíacas/diagnóstico , Arritmias Cardíacas/etnología , Arritmias Cardíacas/terapia , COVID-19/diagnóstico , COVID-19/etnología , COVID-19/terapia , Causas de Muerte , Comorbilidad , Electrocardiografía , Femenino , Factores de Riesgo de Enfermedad Cardiaca , Mortalidad Hospitalaria/etnología , Hospitalización , Humanos , Masculino , Persona de Mediana Edad , Ciudad de Nueva York/epidemiología , Pronóstico , Factores Raciales , Estudios Retrospectivos , Medición de Riesgo , Factores de Tiempo
8.
J Cardiovasc Electrophysiol ; 31(6): 1249-1254, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32281214

RESUMEN

A global coronavirus (COVID-19) pandemic occurred at the start of 2020 and is already responsible for more than 74 000 deaths worldwide, just over 100 years after the influenza pandemic of 1918. At the center of the crisis is the highly infectious and deadly SARS-CoV-2, which has altered everything from individual daily lives to the global economy and our collective consciousness. Aside from the pulmonary manifestations of disease, there are likely to be several electrophysiologic (EP) sequelae of COVID-19 infection and its treatment, due to consequences of myocarditis and the use of QT-prolonging drugs. Most crucially, the surge in COVID-19 positive patients that have already overwhelmed the New York City hospital system requires conservation of hospital resources including personal protective equipment (PPE), reassignment of personnel, and reorganization of institutions, including the EP laboratory. In this proposal, we detail the specific protocol changes that our EP department has adopted during the COVID-19 pandemic, including performance of only urgent/emergent procedures, after hours/7-day per week laboratory operation, single attending-only cases to preserve PPE, appropriate use of PPE, telemedicine and video chat follow-up appointments, and daily conferences to collectively manage the clinical and ethical dilemmas to come. We discuss also discuss how we perform EP procedures on presumed COVID positive and COVID tested positive patients to highlight issues that others in the EP community may soon face in their own institution as the virus continues to spread nationally and internationally.


Asunto(s)
Centros Médicos Académicos/provisión & distribución , Betacoronavirus , Infecciones por Coronavirus/diagnóstico , Electrofisiología/métodos , Equipo de Protección Personal/normas , Neumonía Viral/diagnóstico , COVID-19 , Humanos , Pandemias , SARS-CoV-2
9.
Indian Pacing Electrophysiol J ; 20(6): 250-256, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32861812

RESUMEN

BACKGROUND: The COVID-19 pandemic has greatly altered the practice of cardiac electrophysiology around the world for the foreseeable future. Professional organizations have provided guidance for practitioners, but real-world examples of the consults and responsibilities cardiac electrophysiologists face during a surge of COVID-19 patients is lacking. METHODS: In this observational case series we report on 29 consecutive inpatient electrophysiology consultations at a major academic medical center in New York City, the epicenter of the pandemic in the United States, during a 2 week period from March 30-April 12, 2020, when 80% of hospital beds were occupied by COVID-19 patients, and the New York City metropolitan area accounted for 10% of COVID-19 cases worldwide. RESULTS: Reasons for consultation included: Atrial tachyarrhythmia (31%), cardiac implantable electronic device management (28%), bradycardia (14%), QTc prolongation (10%), ventricular arrhythmia (7%), post-transcatheter aortic valve replacement conduction abnormality (3.5%), ventricular pre-excitation (3.5%), and paroxysmal supraventricular tachycardia (3.5%). Twenty-four patients (86%) were positive for COVID-19 by nasopharyngeal swab. All elective procedures were canceled, and only one urgent device implantation was performed. Thirteen patients (45%) required in-person evaluation and the remainder were managed remotely. CONCLUSION: Our experience shows that the application of a massive alteration in workflow and personnel forced by the pandemic allowed our team to efficiently address the intersection of COVID-19 with a range of electrophysiology issues. This experience will prove useful as guidance for emerging hot spots or areas affected by future waves of the pandemic.

10.
Hum Mol Genet ; 26(2): 333-343, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28069793

RESUMEN

Cardiomyopathy caused by lamin A/C gene (LMNA) mutations (hereafter referred as LMNA cardiomyopathy) is characterized by cardiac conduction abnormalities and left ventricular systolic dysfunction predisposing to heart failure. Previous cardiac transcriptional profiling of LmnaH222P/H222P mouse, a small animal model of LMNA cardiomyopathy, suggested decreased WNT/ß-catenin signalling. We confirmed decreased WNT/ß-catenin signalling in the hearts of these mice by demonstrating decreased ß-catenin and WNT proteins. This was correlated with increased expression of soluble Frizzled-related proteins that modulate the WNT/ß-catenin signalling pathway. Hearts of LmnaH222P/H222P mice also demonstrated lowered expression of the gap junction connexin 43. Activation of WNT/ß-catenin activity with 6-bromoindirubin-3'-oxime improved cardiac contractility and ameliorated intraventricular conduction defects in LmnaH222P/H222P mice, which was associated with increased expression of myocardial connexin 43. These results indicate that decreased WNT/ß-catenin contributes to the pathophysiology of LMNA cardiomyopathy and that drugs activating ß-catenin may be beneficial in affected individuals.


Asunto(s)
Cardiomiopatía Dilatada/genética , Conexina 43/genética , Lamina Tipo A/genética , beta Catenina/genética , Animales , Cardiomiopatía Dilatada/tratamiento farmacológico , Cardiomiopatía Dilatada/fisiopatología , Conexina 43/biosíntesis , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/efectos de los fármacos , Glicoproteínas/biosíntesis , Glicoproteínas/genética , Insuficiencia Cardíaca/tratamiento farmacológico , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/fisiopatología , Humanos , Indoles/administración & dosificación , Péptidos y Proteínas de Señalización Intracelular , Ratones , Mutación , Oximas/administración & dosificación , Disfunción Ventricular Izquierda/tratamiento farmacológico , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/fisiopatología , Proteínas Wnt/genética , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/biosíntesis
11.
Hum Mol Genet ; 25(11): 2220-2233, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27131347

RESUMEN

Cardiomyopathy caused by lamin A/C gene mutations (LMNA cardiomyopathy) is characterized by increased myocardial fibrosis, which impairs left ventricular relaxation and predisposes to heart failure, and cardiac conduction abnormalities. While we previously discovered abnormally elevated extracellular signal-regulated kinase 1/2 (ERK1/2) activities in heart in LMNA cardiomyopathy, its role on the development of myocardial fibrosis remains unclear. We now showed that transforming growth factor (TGF)-ß/Smad signaling participates in the activation of ERK1/2 signaling in LMNA cardiomyopathy. ERK1/2 acts on connective tissue growth factor (CTGF/CCN2) expression to mediate the myocardial fibrosis and left ventricular dysfunction. Studies in vivo demonstrate that inhibiting CTGF/CCN2 using a specific antibody decreases myocardial fibrosis and improves the left ventricular dysfunction. Together, these findings show that cardiac ERK1/2 activity is modulated in part by TGF-ß/Smad signaling, leading to altered activation of CTGF/CCN2 to mediate fibrosis and alter cardiac function. This identifies a novel mechanism in the development of LMNA cardiomyopathy.


Asunto(s)
Cardiomiopatías/genética , Factor de Crecimiento del Tejido Conjuntivo/genética , Fibrosis/genética , Lamina Tipo A/genética , Factor de Crecimiento Transformador beta/genética , Animales , Cardiomiopatías/patología , Fibrosis/patología , Humanos , Sistema de Señalización de MAP Quinasas/genética , Ratones , Ratones Noqueados , Miocardio/metabolismo , Miocardio/patología , Proteínas Smad/genética , Disfunción Ventricular Izquierda/genética , Disfunción Ventricular Izquierda/patología
12.
J Mol Cell Cardiol ; 87: 171-9, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26316303

RESUMEN

BACKGROUND: Mobilization of the innate immune response to clear and metabolize necrotic and apoptotic cardiomyocytes is a prerequisite to heart repair after cardiac injury. Suboptimal kinetics of dying myocyte clearance leads to secondary necrosis, and in the case of the heart, increased potential for collateral loss of neighboring non-regenerative myocytes. Despite the importance of myocyte phagocytic clearance during heart repair, surprisingly little is known about its underlying cell and molecular biology. OBJECTIVE: To determine if phagocytic receptor MERTK is expressed in human hearts and to elucidate key sequential steps and phagocytosis efficiency of dying adult cardiomyocytes, by macrophages. RESULTS: In infarcted human hearts, expression profiles of the phagocytic receptor MER-tyrosine kinase (MERTK) mimicked that found in experimental ischemic mouse hearts. Electron micrographs of myocardium identified MERTK signal along macrophage phagocytic cups and Mertk-/- macrophages contained reduced digested myocyte debris after myocardial infarction. Ex vivo co-culture of primary macrophages and adult cardiomyocyte apoptotic bodies revealed reduced engulfment relative to resident cardiac fibroblasts. Inefficient clearance was not due to the larger size of myocyte apoptotic bodies, nor were other key steps preceding the formation of phagocytic synapses significantly affected; this included macrophage chemotaxis and direct binding of phagocytes to myocytes. Instead, suppressed phagocytosis was directly associated with myocyte-induced inactivation of MERTK, which was partially rescued by genetic deletion of a MERTK proteolytic susceptibility site. CONCLUSION: Utilizing an ex vivo co-cultivation approach to model key cellular and molecular events found in vivo during infarction, cardiomyocyte phagocytosis was found to be inefficient, in part due to myocyte-induced shedding of macrophage MERTK. These findings warrant future studies to identify other cofactors of macrophage-cardiomyocyte cross-talk that contribute to cardiac pathophysiology.


Asunto(s)
Inmunidad Innata/genética , Infarto del Miocardio/genética , Miocitos Cardíacos/metabolismo , Fagocitosis/genética , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Animales , Apoptosis/genética , Apoptosis/inmunología , Línea Celular , Técnicas de Cocultivo , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Ratones , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Miocitos Cardíacos/patología , Necrosis/genética , Necrosis/metabolismo , Fagocitosis/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/genética , Tirosina Quinasa c-Mer
13.
J Pharmacol Exp Ther ; 355(3): 397-409, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26407721

RESUMEN

Mutations in the leucine-rich repeat kinase 2 (LRRK2) gene are the most common genetic cause of familial and sporadic Parkinson's disease (PD). That the most prevalent mutation, G2019S, leads to increased kinase activity has led to a concerted effort to identify LRRK2 kinase inhibitors as a potential disease-modifying therapy for PD. An internal medicinal chemistry effort identified several potent and highly selective compounds with favorable drug-like properties. Here, we characterize the pharmacological properties of cis-2,6-dimethyl-4-(6-(5-(1-methylcyclopropoxy)-1H-indazol-3-yl)pyrimidin-4-yl)morpholine (MLi-2), a structurally novel, highly potent, and selective LRRK2 kinase inhibitor with central nervous system activity. MLi-2 exhibits exceptional potency in a purified LRRK2 kinase assay in vitro (IC50 = 0.76 nM), a cellular assay monitoring dephosphorylation of LRRK2 pSer935 LRRK2 (IC50 = 1.4 nM), and a radioligand competition binding assay (IC50 = 3.4 nM). MLi-2 has greater than 295-fold selectivity for over 300 kinases in addition to a diverse panel of receptors and ion channels. Acute oral and subchronic dosing in MLi-2 mice resulted in dose-dependent central and peripheral target inhibition over a 24-hour period as measured by dephosphorylation of pSer935 LRRK2. Treatment of MitoPark mice with MLi-2 was well tolerated over a 15-week period at brain and plasma exposures >100× the in vivo plasma IC50 for LRRK2 kinase inhibition as measured by pSer935 dephosphorylation. Morphologic changes in the lung, consistent with enlarged type II pneumocytes, were observed in MLi-2-treated MitoPark mice. These data demonstrate the suitability of MLi-2 as a compound to explore LRRK2 biology in cellular and animal models.


Asunto(s)
Antiparkinsonianos/efectos adversos , Antiparkinsonianos/uso terapéutico , Indazoles/farmacología , Inhibidores de Proteínas Quinasas/efectos adversos , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/patología , Animales , Conducta Animal/efectos de los fármacos , Unión Competitiva , Encéfalo/metabolismo , Química Encefálica/efectos de los fármacos , Línea Celular , Corteza Cerebral/efectos de los fármacos , Corteza Cerebral/metabolismo , Relación Dosis-Respuesta a Droga , Humanos , Proteína 2 Quinasa Serina-Treonina Rica en Repeticiones de Leucina , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Mutación/genética , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/patología , Enfermedad de Parkinson/psicología , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo
14.
Circ Res ; 113(7): 871-80, 2013 Sep 13.
Artículo en Inglés | MEDLINE | ID: mdl-23825359

RESUMEN

RATIONALE: Sympathetic nervous system triggered activation of protein kinase A, which phosphorylates several targets within cardiomyocytes, augments inotropy, chronotropy, and lusitropy. An important target of ß-adrenergic stimulation is the sarcolemmal L-type Ca(2+) channel, CaV1.2, which plays a key role in cardiac excitation-contraction coupling. The molecular mechanisms of ß-adrenergic regulation of CaV1.2 in cardiomyocytes, however, are incompletely known. Recently, it has been postulated that proteolytic cleavage at Ala(1800) and protein kinase A phosphorylation of Ser(1700) are required for ß-adrenergic modulation of CaV1.2. OBJECTIVE: To assess the role of Ala(1800) in the cleavage of α1C and the role of Ser(1700) and Thr(1704) in mediating the adrenergic regulation of CaV1.2 in the heart. METHODS AND RESULTS: Using a transgenic approach that enables selective and inducible expression in mice of FLAG-epitope-tagged, dihydropyridine-resistant CaV1.2 channels harboring mutations at key regulatory sites, we show that adrenergic regulation of CaV1.2 current and fractional shortening of cardiomyocytes do not require phosphorylation of either Ser(1700) or Thr(1704) of the α1C subunit. The presence of Ala(1800) and the (1798)NNAN(1801) motif in α1C is not required for proteolytic cleavage of the α1C C-terminus, and deletion of these residues did not perturb adrenergic modulation of CaV1.2 current. CONCLUSIONS: These results show that protein kinase A phosphorylation of α1C Ser(1700) does not have a major role in the sympathetic stimulation of Ca(2+) current and contraction in the adult murine heart. Moreover, this new transgenic approach enables functional and reproducible screening of α1C mutants in freshly isolated adult cardiomyocytes in a reliable, timely, cost-effective manner.


Asunto(s)
Adrenérgicos/farmacología , Canales de Calcio Tipo L/metabolismo , Miocitos Cardíacos/metabolismo , Potenciales de Acción/efectos de los fármacos , Alanina/genética , Alanina/metabolismo , Secuencias de Aminoácidos , Animales , Canales de Calcio Tipo L/química , Canales de Calcio Tipo L/genética , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Ratones , Mutación , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/fisiología , Fosforilación , Proteolisis , Conejos , Serina/genética , Serina/metabolismo , Treonina/genética , Treonina/metabolismo
15.
Circ Res ; 113(8): 1004-12, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23836795

RESUMEN

RATIONALE: Efficient clearance of apoptotic cells (efferocytosis) is a prerequisite for inflammation resolution and tissue repair. After myocardial infarction, phagocytes are recruited to the heart and promote clearance of dying cardiomyocytes. The molecular mechanisms of efferocytosis of cardiomyocytes and in the myocardium are unknown. The injured heart provides a unique model to examine relationships between efferocytosis and subsequent inflammation resolution, tissue remodeling, and organ function. OBJECTIVE: We set out to identify mechanisms of dying cardiomyocyte engulfment by phagocytes and, for the first time, to assess the causal significance of disrupting efferocytosis during myocardial infarction. METHODS AND RESULTS: In contrast to other apoptotic cell receptors, macrophage myeloid-epithelial-reproductive tyrosine kinase was necessary and sufficient for efferocytosis of cardiomyocytes ex vivo. In mice, Mertk was specifically induced in Ly6c(LO) myocardial phagocytes after experimental coronary occlusion. Mertk deficiency led to an accumulation of apoptotic cardiomyocytes, independently of changes in noncardiomyocytes, and a reduced index of in vivo efferocytosis. Importantly, suppressed efferocytosis preceded increases in myocardial infarct size and led to delayed inflammation resolution and reduced systolic performance. Reduced cardiac function was reproduced in chimeric mice deficient in bone marrow Mertk; reciprocal transplantation of Mertk(+/+) marrow into Mertk(-/-) mice corrected systolic dysfunction. Interestingly, an inactivated form of myeloid-epithelial-reproductive tyrosine kinase, known as solMER, was identified in infarcted myocardium, implicating a natural mechanism of myeloid-epithelial-reproductive tyrosine kinase inactivation after myocardial infarction. CONCLUSIONS: These data collectively and directly link efferocytosis to wound healing in the heart and identify Mertk as a significant link between acute inflammation resolution and organ function.


Asunto(s)
Apoptosis , Inflamación/enzimología , Macrófagos/enzimología , Infarto del Miocardio/enzimología , Miocitos Cardíacos/enzimología , Fagocitosis , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Cicatrización de Heridas , Animales , Antígenos Ly/metabolismo , Trasplante de Médula Ósea , Antígenos CD36/deficiencia , Antígenos CD36/genética , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Contracción Miocárdica , Infarto del Miocardio/genética , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/fisiopatología , Miocitos Cardíacos/inmunología , Miocitos Cardíacos/patología , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Tirosina Quinasas Receptoras/deficiencia , Proteínas Tirosina Quinasas Receptoras/genética , Recuperación de la Función , Transducción de Señal , Factores de Tiempo , Quimera por Trasplante , Función Ventricular Izquierda , Remodelación Ventricular , Tirosina Quinasa c-Mer
16.
J Mol Cell Cardiol ; 75: 58-63, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25008120

RESUMEN

Pathologic cardiac hypertrophy can lead to heart failure, but the mechanisms involved are poorly understood. SERCA2 is critical for normal cardiac calcium handling and function and SERCA2 mRNA and protein levels are reduced by cardiac hypertrophy. We hypothesized that extracellular signal-regulated kinase (ERK) 1/2 activation during hypertrophy reduced SERCA2 transcription. Using a neonatal rat ventricular myocyte model of hypertrophy, we found that pharmacologic inhibitors of ERK activation preserve SERCA2 mRNA levels during hypertrophy. ERK activation is sufficient to reduce SERCA2 mRNA. We determined that ERK represses SERCA2 transcription via nuclear factor-kappaB (NFkB), and activation of NFkB is sufficient to reduce SERCA2 mRNA in cardiomyocytes. This work establishes novel connections between ERK, NFkB, and SERCA2 repression during cardiac hypertrophy. This mechanism may have implications for the progression of hypertrophy to heart failure.


Asunto(s)
Cardiomegalia/enzimología , Cardiomegalia/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/genética , Transcripción Genética , Animales , Animales Recién Nacidos , Regulación de la Expresión Génica , Humanos , Ratones , Modelos Biológicos , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/patología , FN-kappa B/metabolismo , Fenilefrina , Regiones Promotoras Genéticas/genética , Unión Proteica/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , ATPasas Transportadoras de Calcio del Retículo Sarcoplásmico/metabolismo
17.
Hum Mol Genet ; 21(19): 4325-33, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22773734

RESUMEN

We previously interrogated the transcriptome in heart tissue from Lmna(H222P/H222P) mice, a mouse model of cardiomyopathy caused by lamin A/C gene (LMNA) mutation, and found that the extracellular signal-regulated kinase 1/2 and Jun N-terminal kinase branches of the mitogen-activated protein (MAP) kinase signaling pathway were abnormally hyperactivated prior to the onset of significant cardiac impairment. We have now used an alternative gene expression analysis tool to reanalyze this transcriptome and identify hyperactivation of a third branch of the MAP kinase cascade, p38α signaling. Biochemical analysis of hearts from Lmna(H222P/H222P) mice showed enhanced p38α activation prior to and after the onset of heart disease as well as in hearts from human subjects with cardiomyopathy caused by LMNA mutations. Treatment of Lmna(H222P/H222P) mice with the p38α inhibitor ARRY-371797 prevented left ventricular dilatation and deterioration of fractional shortening compared with placebo-treated mice but did not block the expression of collagen genes involved in cardiac fibrosis. These results demonstrate that three different branches of the MAP kinase signaling pathway with overlapping consequences are involved in the pathogenesis of cardiomyopathy caused by LMNA mutations. They further suggest that pharmacological inhibition of p38α may be useful in the treatment of this disease.


Asunto(s)
Cardiomiopatía Dilatada/enzimología , Etilenodiaminas/farmacología , Indazoles/farmacología , Lamina Tipo A/genética , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Mutación Missense , Transducción de Señal , Adolescente , Animales , Cardiomiopatía Dilatada/genética , Cardiomiopatía Dilatada/metabolismo , Células Cultivadas , Femenino , Humanos , Lamina Tipo A/metabolismo , Masculino , Ratones , Ratones Transgénicos , Persona de Mediana Edad , Proteína Quinasa 14 Activada por Mitógenos/genética , Miocitos Cardíacos/enzimología , Miocitos Cardíacos/metabolismo , Adulto Joven
18.
J Mol Cell Cardiol ; 59: 151-8, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23517696

RESUMEN

In humans, obesity is associated with long QT, increased frequency of premature ventricular complexes, and sudden cardiac death. The mechanisms of the pro-arrhythmic electrophysiologic remodeling of obesity are poorly understood. We tested the hypothesis that there is decreased expression of voltage-gated potassium channels in the obese heart, leading to long QT. Using implanted telemeters, we found that diet-induced obese (DIO) wild-type mice have impaired cardiac repolarization, demonstrated by long QT, as well as more frequent ventricular ectopy, similar to obese humans. DIO mice have reduced protein and mRNA levels of the potassium channel Kv1.5 caused by a reduction of the transcription factor cyclic AMP response element binding protein (CREB) in DIO hearts. We found that CREB knock-down by siRNA reduces Kv1.5, CREB binds to the Kv1.5 promoter in the heart, and CREB increases transcription of mouse and human Kv1.5 promoters. The reduction in CREB protein during lipotoxicity can be rescued by inhibiting protein kinase D (PKD). Our results identify a mechanism for obesity-induced electrophysiologic remodeling in the heart, namely PKD-induced reduction of CREB, which in turn decreases expression of the potassium channel Kv1.5.


Asunto(s)
Arritmias Cardíacas/etiología , Arritmias Cardíacas/metabolismo , Dieta Alta en Grasa/efectos adversos , Síndrome de QT Prolongado/etiología , Síndrome de QT Prolongado/metabolismo , Obesidad/complicaciones , Obesidad/etiología , Obesidad/metabolismo , Canales de Potasio con Entrada de Voltaje/metabolismo , Animales , Línea Celular , Inmunoprecipitación de Cromatina , Proteína de Unión a Elemento de Respuesta al AMP Cíclico , Immunoblotting , Canal de Potasio Kv1.5/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína Quinasa C/metabolismo , Ratas , Reacción en Cadena en Tiempo Real de la Polimerasa
19.
N C Med J ; 74(4): 330-3, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24044155

RESUMEN

The need to improve population health is critical. This commentary explores how the Patient Protection and Affordable Care Act of 2010 (ACA) can help us improve population health, highlights some of the actions North Carolina has taken in response to the ACA's provisions, and discusses the value of health investments in the future.


Asunto(s)
Estado de Salud , Patient Protection and Affordable Care Act , Humanos , Evaluación de Necesidades , North Carolina , Prevención Primaria , Salud Pública , Estados Unidos
20.
Circulation ; 124(25): 2812-21, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22124376

RESUMEN

BACKGROUND: Diabetes mellitus and obesity, which confer an increased risk of sudden cardiac death, are associated with cardiomyocyte lipid accumulation and altered cardiac electric properties, manifested by prolongation of the QRS duration and QT interval. It is difficult to distinguish the contribution of cardiomyocyte lipid accumulation from the contribution of global metabolic defects to the increased incidence of sudden death and electric abnormalities. METHODS AND RESULTS: In order to study the effects of metabolic abnormalities on arrhythmias without the complex systemic effects of diabetes mellitus and obesity, we studied transgenic mice with cardiac-specific overexpression of peroxisome proliferator-activated receptor γ 1 (PPARγ1) via the cardiac α-myosin heavy-chain promoter. The PPARγ transgenic mice develop abnormal accumulation of intracellular lipids and die as young adults before any significant reduction in systolic function. Using implantable ECG telemeters, we found that these mice have prolongation of the QRS and QT intervals and spontaneous ventricular arrhythmias, including polymorphic ventricular tachycardia and ventricular fibrillation. Isolated cardiomyocytes demonstrated prolonged action potential duration caused by reduced expression and function of the potassium channels responsible for repolarization. Short-term exposure to pioglitazone, a PPARγ agonist, had no effect on mortality or rhythm in WT mice but further exacerbated the arrhythmic phenotype and increased the mortality in the PPARγ transgenic mice. CONCLUSIONS: Our findings support an important link between PPARγ activation, cardiomyocyte lipid accumulation, ion channel remodeling, and increased cardiac mortality.


Asunto(s)
PPAR gamma/genética , Periodo Refractario Electrofisiológico/fisiología , Taquicardia Ventricular/fisiopatología , Fibrilación Ventricular/fisiopatología , Potenciales de Acción/fisiología , Animales , Calcio/metabolismo , Conexina 43/genética , Conexina 43/metabolismo , Muerte Súbita Cardíaca/epidemiología , Modelos Animales de Enfermedad , Electrocardiografía , Hipoglucemiantes/farmacología , Incidencia , Lípido A/metabolismo , Ratones , Ratones Transgénicos , Miocardio/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/fisiología , PPAR gamma/fisiología , Fenotipo , Pioglitazona , Potasio/metabolismo , Canales de Potasio con Entrada de Voltaje/genética , Canales de Potasio con Entrada de Voltaje/fisiología , Periodo Refractario Electrofisiológico/efectos de los fármacos , Sodio/metabolismo , Taquicardia Ventricular/genética , Taquicardia Ventricular/mortalidad , Tiazolidinedionas/farmacología , Fibrilación Ventricular/genética , Fibrilación Ventricular/mortalidad , Remodelación Ventricular/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA