Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 250
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
South Med J ; 117(2): 67-71, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38307500

RESUMEN

Without rural hospitals, many patients may not have access to essential services, or even any health care. Rural hospitals provide a community hub for local access to primary care and emergency services, as well as a bridge to specialized care outside the community. The goal of this review was to demonstrate how the University of Arkansas for Medical Sciences supports and empowers rural hospitals through an alliance that provides cost savings through clinical networks, collaborative purchasing, and leveraged services; workforce recruitment and education; telemedicine and distance learning; community outreach; and access to best practices, resources, and tools for hospital transformation. Born out of grassroots efforts in the rural US South, this model alliance, the Arkansas Rural Health Partnership, with the University of Arkansas for Medical Sciences supporting as an academic medical center participant, offers resources and programs intended to help rural hospitals and healthcare providers survive and even thrive in the challenging landscape that is forcing many other rural hospitals to close. The Arkansas Rural Health Partnership model is relevant for rural states that are seeking to develop or reenvision rural hospital alliances with academic medical centers to the benefit of the hospitals and the health of their communities and state.


Asunto(s)
Servicios de Salud Rural , Telemedicina , Humanos , Hospitales Rurales , Atención a la Salud , Salud Rural , Arkansas , Población Rural
2.
PLoS Genet ; 14(9): e1007664, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30222779

RESUMEN

CHIP (carboxyl terminus of heat shock 70-interacting protein) has long been recognized as an active member of the cellular protein quality control system given the ability of CHIP to function as both a co-chaperone and ubiquitin ligase. We discovered a genetic disease, now known as spinocerebellar autosomal recessive 16 (SCAR16), resulting from a coding mutation that caused a loss of CHIP ubiquitin ligase function. The initial mutation describing SCAR16 was a missense mutation in the ubiquitin ligase domain of CHIP (p.T246M). Using multiple biophysical and cellular approaches, we demonstrated that T246M mutation results in structural disorganization and misfolding of the CHIP U-box domain, promoting oligomerization, and increased proteasome-dependent turnover. CHIP-T246M has no ligase activity, but maintains interactions with chaperones and chaperone-related functions. To establish preclinical models of SCAR16, we engineered T246M at the endogenous locus in both mice and rats. Animals homozygous for T246M had both cognitive and motor cerebellar dysfunction distinct from those observed in the CHIP null animal model, as well as deficits in learning and memory, reflective of the cognitive deficits reported in SCAR16 patients. We conclude that the T246M mutation is not equivalent to the total loss of CHIP, supporting the concept that disease-causing CHIP mutations have different biophysical and functional repercussions on CHIP function that may directly correlate to the spectrum of clinical phenotypes observed in SCAR16 patients. Our findings both further expand our basic understanding of CHIP biology and provide meaningful mechanistic insight underlying the molecular drivers of SCAR16 disease pathology, which may be used to inform the development of novel therapeutics for this devastating disease.


Asunto(s)
Cognición , Actividad Motora/genética , Dominios Proteicos/genética , Ataxias Espinocerebelosas/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Conducta Animal , Sistemas CRISPR-Cas/genética , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Fenotipo , Mutación Puntual , Multimerización de Proteína/genética , Ratas , Ratas Sprague-Dawley , Ataxias Espinocerebelosas/congénito , Ubiquitina-Proteína Ligasas/metabolismo
3.
J Biol Chem ; 294(50): 19236-19245, 2019 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-31619515

RESUMEN

Monogenetic disorders that cause cerebellar ataxia are characterized by defects in gait and atrophy of the cerebellum; however, patients often suffer from a spectrum of disease, complicating treatment options. Spinocerebellar ataxia autosomal recessive 16 (SCAR16) is caused by coding mutations in STUB1, a gene that encodes the multifunctional enzyme CHIP (C terminus of HSC70-interacting protein). The disease spectrum of SCAR16 includes a varying age of disease onset, cognitive dysfunction, increased tendon reflex, and hypogonadism. Although SCAR16 mutations span the multiple functional domains of CHIP, it is unclear whether the location of the mutation and the change in the biochemical properties of CHIP contributes to the clinical spectrum of SCAR16. In this study, we examined relationships between the clinical phenotypes of SCAR16 patients and the changes in biophysical, biochemical, and functional properties of the corresponding mutated protein. We found that the severity of ataxia did not correlate with age of onset; however, cognitive dysfunction, increased tendon reflex, and ancestry were able to predict 54% of the variation in ataxia severity. We further identified domain-specific relationships between biochemical changes in CHIP and clinical phenotypes and specific biochemical activities that associate selectively with either increased tendon reflex or cognitive dysfunction, suggesting that specific changes to CHIP-HSC70 dynamics contribute to the clinical spectrum of SCAR16. Finally, linear models of SCAR16 as a function of the biochemical properties of CHIP support the concept that further inhibiting mutant CHIP activity lessens disease severity and may be useful in the design of patient-specific targeted approaches to treat SCAR16.


Asunto(s)
Proteínas del Choque Térmico HSC70/metabolismo , Trastornos del Neurodesarrollo/metabolismo , Ataxias Espinocerebelosas/metabolismo , Proteínas del Choque Térmico HSC70/genética , Humanos , Método de Montecarlo , Análisis Multivariante , Mutación , Trastornos del Neurodesarrollo/genética , Fenotipo , Ataxias Espinocerebelosas/genética
4.
Circ Res ; 123(4): 477-494, 2018 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-30355249

RESUMEN

Our understanding of the role of the vascular endothelium has evolved over the past 2 decades, with the recognition that it is a dynamically regulated organ and that it plays a nodal role in a variety of physiological and pathological processes. Endothelial cells (ECs) are not only a barrier between the circulation and peripheral tissues, but also actively regulate vascular tone, blood flow, and platelet function. Dysregulation of ECs contributes to pathological conditions such as vascular inflammation, atherosclerosis, hypertension, cardiomyopathy, retinopathy, neuropathy, and cancer. The close anatomic relationship between vascular endothelium and highly vascularized metabolic organs/tissues suggests that the crosstalk between ECs and these organs is vital for both vascular and metabolic homeostasis. Numerous reports support that hyperlipidemia, hyperglycemia, and other metabolic stresses result in endothelial dysfunction and vascular complications. However, how ECs may regulate metabolic homeostasis remains poorly understood. Emerging data suggest that the vascular endothelium plays an unexpected role in the regulation of metabolic homeostasis and that endothelial dysregulation directly contributes to the development of metabolic disorders. Here, we review recent studies about the pivotal role of ECs in glucose and lipid homeostasis. In particular, we introduce the concept that the endothelium adjusts its barrier function to control the transendothelial transport of fatty acids, lipoproteins, LPLs (lipoprotein lipases), glucose, and insulin. In addition, we summarize reports that ECs communicate with metabolic cells through EC-secreted factors and we discuss how endothelial dysregulation contributes directly to the development of obesity, insulin resistance, dyslipidemia, diabetes mellitus, cognitive defects, and fatty liver disease.


Asunto(s)
Endotelio Vascular/metabolismo , Metabolismo Energético , Homeostasis , Enfermedades Metabólicas/etiología , Animales , Humanos
5.
Circulation ; 138(22): 2569-2575, 2018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30571349

RESUMEN

Artificial intelligence offers the potential for transformational advancement in cardiovascular care delivery, yet practical applications of this technology have yet to be embedded in clinical workflows and systems. Recent advances in machine learning algorithms and accessibility to big data sources have created the ability for software to solve highly specialized problems outside of health care, such as autonomous driving, speech recognition, and game playing (chess and Go), at superhuman efficiency previously not thought possible. To date, high-order cognitive problems in cardiovascular research such as differential diagnosis, treatment options, and clinical risk stratification have been difficult to address at scale with artificial intelligence. The practical application of artificial intelligence in the underlying operational processes in the delivery of cardiac care may be more amenable where adoption has great potential to fundamentally transform care delivery while maintaining the core quality and service that our patients demand. In this article, we provide an overview on how these artificial intelligence platforms can be implemented to improve the operational delivery of care for patients with cardiovascular disease.


Asunto(s)
Atención a la Salud/métodos , Aprendizaje Automático , Algoritmos , Enfermedades Cardiovasculares/diagnóstico , Enfermedades Cardiovasculares/diagnóstico por imagen , Enfermedades Cardiovasculares/terapia , Humanos , Imagen por Resonancia Magnética
6.
Am J Pathol ; 188(7): 1676-1692, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29758183

RESUMEN

The muscle-specific ubiquitin ligase atrogin-1 (MAFbx) has been identified as a critical regulator of pathologic and physiological cardiac hypertrophy; it regulates these processes by ubiquitinating transcription factors [nuclear factor of activated T-cells and forkhead box O (FoxO) 1/3]. However, the role of atrogin-1 in regulating transcription factors in aging has not previously been described. Atrogin-1 cardiomyocyte-specific transgenic (Tg+) adult mice (α-major histocompatibility complex promoter driven) have normal cardiac function and size. Herein, we demonstrate that 18-month-old atrogin-1 Tg+ hearts exhibit significantly increased anterior wall thickness without functional impairment versus wild-type mice. Histologic analysis at 18 months revealed atrogin-1 Tg+ mice had significantly less fibrosis and significantly greater nuclei and cardiomyocyte cross-sectional analysis. Furthermore, by real-time quantitative PCR, atrogin-1 Tg+ had increased Col 6a4, 6a5, 6a6, matrix metalloproteinase 8 (Mmp8), and Mmp9 mRNA, suggesting a role for atrogin-1 in regulating collagen deposits and MMP-8 and MMP-9. Because atrogin-1 Tg+ mice exhibited significantly less collagen deposition and protein levels, enhanced Mmp8 and Mmp9 mRNA may offer one mechanism by which collagen levels are kept in check in the aged atrogin-1 Tg+ heart. In addition, atrogin-1 Tg+ hearts showed enhanced FoxO1/3 activity. The present study shows a novel link between atrogin-1-mediated regulation of FoxO1/3 activity and reduced collagen deposition and fibrosis in the aged heart. Therefore, targeting FoxO1/3 activity via the muscle-specific atrogin-1 ubiquitin ligase may offer a muscle-specific method to modulate aging-related cardiac fibrosis.


Asunto(s)
Envejecimiento , Cardiomegalia/prevención & control , Fenómenos Fisiológicos Cardiovasculares , Fibrosis/prevención & control , Proteínas Musculares/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Animales , Cardiomegalia/etiología , Cardiomegalia/metabolismo , Estudios Transversales , Fibrosis/etiología , Fibrosis/metabolismo , Ratones , Ratones Transgénicos , Proteínas Musculares/genética , Proteínas Ligasas SKP Cullina F-box/genética , Transducción de Señal
7.
J Am Soc Nephrol ; 29(3): 936-948, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29242247

RESUMEN

The importance of the kidney distal convoluted tubule (DCT) and cortical collecting duct (CCD) is highlighted by various water and electrolyte disorders that arise when the unique transport properties of these segments are disturbed. Despite this critical role, little is known about which proteins have a regulatory role in these cells and how these cells can be regulated by individual physiologic stimuli. By combining proteomics, bioinformatics, and cell biology approaches, we found that the E3 ubiquitin ligase CHIP is highly expressed throughout the collecting duct; is modulated in abundance by vasopressin; interacts with aquaporin-2 (AQP2), Hsp70, and Hsc70; and can directly ubiquitylate the water channel AQP2 in vitro shRNA knockdown of CHIP in CCD cells increased AQP2 protein t1/2 and reduced AQP2 ubiquitylation, resulting in greater levels of AQP2 and phosphorylated AQP2. CHIP knockdown increased the plasma membrane abundance of AQP2 in these cells. Compared with wild-type controls, CHIP knockout mice or novel CRISPR/Cas9 mice without CHIP E3 ligase activity had greater AQP2 abundance and altered renal water handling, with decreased water intake and urine volume, alongside higher urine osmolality. We did not observe significant changes in other water- or sodium-transporting proteins in the gene-modified mice. In summary, these results suggest that CHIP regulates AQP2 and subsequently, renal water handling.


Asunto(s)
Acuaporina 2/metabolismo , Homeostasis/genética , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Agua/metabolismo , Animales , Células Cultivadas , Ontología de Genes , Silenciador del Gen , Proteínas HSP70 de Choque Térmico/metabolismo , Túbulos Renales Colectores/metabolismo , Túbulos Renales Distales/metabolismo , Ratones , Proteómica , Ubiquitinación
8.
Am J Physiol Cell Physiol ; 315(6): C830-C838, 2018 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-30257106

RESUMEN

Hyperphosphatemia, the elevated level of inorganic phosphate (Pi) in serum, is associated with increased cardiovascular morbidities and mortality. The effects of high Pi on endothelial cells are not well studied. This study investigated high Pi-induced endothelial cell apoptosis and the role of microRNA-21. Mouse myocardial endothelial cells (MEC) were cultured in normal (1 mM) and high (5 mM) Pi conditions. Apoptosis was detected by TUNEL staining and flow cytometry. MicroRNA profiles of MEC response to changes in Pi concentration were obtained using gene expression arrays. Expression levels of the microRNA-21 target genes, programmed cell death gene 4 ( PDCD4), poly(ADP-ribose) polymerase ( PARP), and phosphatase and tensin homolog ( PTEN), as well as NF-κB were measured by Western blotting and RT-PCR. MicroRNA-21-specific inhibitors and mimics were used to study effects of microRNA-21 on MEC apoptosis and gene expression regulations. High Pi induced MEC apoptosis and upregulated microRNA-21 expression. MicroRNA-21-specific mimics reproduced high Pi-induced apoptosis in normal Pi medium, and microRNA-21 inhibitors ameliorated the high Pi induction of apoptosis, suggesting that microRNA-21 mediated high Pi-induced MEC apoptosis. The microRNA-21 targets PDCD4, PTEN, PARP, and NF-κB were significantly downregulated in high Pi conditions. High Pi-induced downregulation of PDCD4 was abolished by microRNA-21 inhibitors and selective ERK inhibitor (selumetinib) and was reproduced by microRNA-21 mimics. Inhibitors and mimics of microRNA-21 did not have effects on high Pi-induced NF-κB downregulation. Selumetinib blocked high Pi-induced NF-κB downregulation. MicroRNA-21 mediates high Pi-induced endothelial cell apoptosis, which involves an ERK1/2/microRNA-21/PDCD4 pathway. High Pi-induced downregulation of NF-κB expression is mediated by an ERK1/2 signaling-dependent but microRNA-21-independent mechanism.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , MicroARNs/genética , Miocardio/metabolismo , Poli(ADP-Ribosa) Polimerasas/genética , Proteínas de Unión al ARN/genética , Animales , Apoptosis/genética , Bencimidazoles/administración & dosificación , Células Endoteliales/metabolismo , Regulación de la Expresión Génica/genética , Humanos , Hiperfosfatemia/sangre , Hiperfosfatemia/patología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Miocardio/patología , FN-kappa B/genética , Fosfohidrolasa PTEN/genética , Fosfatos/sangre
9.
Am J Pathol ; 187(12): 2895-2911, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29153655

RESUMEN

Our goal was to measure the association of CXCL5 and molecular phenotypes associated with coronary atherosclerosis severity in patients at least 65 years old. CXCL5 is classically defined as a proinflammatory chemokine, but its role in chronic inflammatory diseases, such as coronary atherosclerosis, is not well defined. We enrolled individuals who were at least 65 years old and undergoing diagnostic cardiac catheterization. Coronary artery disease (CAD) severity was quantified in each subject via coronary angiography by calculating a CAD score. Circulating CXCL5 levels were measured from plasma, and both DNA genotyping and mRNA expression levels in peripheral blood mononuclear cells were quantified via microarray gene chips. We observed a negative association of CXCL5 levels with CAD at an odds ratio (OR) of 0.46 (95% CI, 0.27-0.75). Controlling for covariates, including sex, statin use, hypertension, hyperlipidemia, obesity, self-reported race, smoking, and diabetes, the OR was not significantly affected [OR, 0.54 (95% CI, 0.31-0.96)], consistent with a protective role for CXCL5 in coronary atherosclerosis. We also identified 18 genomic regions with expression quantitative trait loci of genes correlated with both CAD severity and circulating CXCL5 levels. Our clinical findings are consistent with the emerging link between chemokines and atherosclerosis and suggest new therapeutic targets for CAD.


Asunto(s)
Quimiocina CXCL5/sangre , Enfermedad de la Arteria Coronaria/sangre , Anciano , Quimiocina CXCL5/genética , Enfermedad de la Arteria Coronaria/genética , Femenino , Humanos , Masculino
10.
Metabolomics ; 14(1): 8, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-30104954

RESUMEN

Introduction: The effects of exercise on the heart and its resistance to disease are well-documented. Recent studies have identified that exercise-induced resistance to arrhythmia is due to the preservation of mitochondrial membrane potential. Objectives: To identify novel metabolic changes that occur parallel to these mitochondrial alterations, we performed non-targeted metabolomics analysis on hearts from sedentary and exercise-trained rats challenged with isolated heart ischemia-reperfusion injury (I/R). Methods: Eight-week old Sprague-Dawley rats were treadmill trained 5 days/week for 6 weeks (exercise duration and intensity progressively increased to 1 h at 30 m/min up a 10.5% incline, 75-80% VO2max). The recovery of pre-ischemic function for sedentary rat hearts was 28.8 ± 5.4% (N = 12) compared to exercise trained hearts, which recovered 51.9% ± 5.7 (N = 14) (p < 0.001). Results: Non-targeted GC-MS metabolomics analysis of (1) sedentary rat hearts; (2) exercise-trained rat hearts; (3) sedentary rat hearts challenged with global ischemia-reperfusion (I/R) injury; and (4) exercise-trained rat hearts challenged with global I/R (10/group) revealed 15 statistically significant metabolites between groups by ANOVA using Metaboanalyst (p < 0.001). Enrichment analysis of these metabolites for pathway-associated metabolic sets indicated a > 10-fold enrichment for ammonia recycling and protein biosynthesis. Subsequent comparison of the sedentary hearts post-I/R and exercise-trained hearts post-I/R further identified significant differences in three metabolites (oleic acid, pantothenic acid, and campesterol) related to pantothenate and CoA biosynthesis (p ≤ 1.24E-05, FDR ≤ 5.07E-4). Conclusions: These studies shed light on novel mechanisms in which exercise-induced cardioprotection occurs in I/R that complement both the mitochondrial stabilization and antioxidant mechanisms recently described. These findings also link protein synthesis and protein degradation (protein quality control mechanisms) with exercise-linked cardioprotection and mitochondrial susceptibility for the first time in cardiac I/R.


Asunto(s)
Potencial de la Membrana Mitocondrial/fisiología , Membranas Mitocondriales/fisiología , Daño por Reperfusión/metabolismo , Animales , Enfermedad de la Arteria Coronaria/metabolismo , Modelos Animales de Enfermedad , Cromatografía de Gases y Espectrometría de Masas/métodos , Corazón/fisiopatología , Isquemia/metabolismo , Masculino , Metaboloma/fisiología , Metabolómica/métodos , Mitocondrias Cardíacas/metabolismo , Isquemia Miocárdica/metabolismo , Reperfusión Miocárdica , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/metabolismo , Condicionamiento Físico Animal/fisiología , Ratas , Ratas Sprague-Dawley , Conducta Sedentaria
11.
Arterioscler Thromb Vasc Biol ; 37(8): 1524-1535, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28596374

RESUMEN

OBJECTIVE: Bacterial endotoxin (lipopolysaccharide)-mediated sepsis involves dysregulated systemic inflammation, which injures the lung and other organs, often fatally. Vascular endothelial cells act as both targets and mediators of lipopolysaccharide-induced inflammatory responses. Dysfunction of endothelium results in increases of proinflammatory cytokine production and permeability leakage. BMPER (bone morphogenetic protein-binding endothelial regulator), an extracellular modulator of bone morphogenetic protein signaling, has been identified as a vital component in chronic endothelial inflammatory responses and atherosclerosis. However, it is unclear whether BMPER also regulates inflammatory response in an acute setting such as sepsis. To address this question, we investigated the role of BMPER during lipopolysaccharide-induced acute lung injury. APPROACH AND RESULTS: Mice missing 1 allele of BMPER (BMPER+/- mice used in the place of BMPER-/- mice that die at birth) were used for lipopolysaccharide challenge. Lipopolysaccharide-induced pulmonary inflammation and injury was reduced in BMPER+/- mice as shown by several measures, including survival rate, infiltration of inflammatory cells, edema, and production of proinflammatory cytokines. Mechanistically, we have demonstrated that BMPER is required and sufficient for the activation of nuclear factor of activated T cells c1. This BMPER-induced nuclear factor of activated T cells activation is coordinated by multiple signaling pathways, including bone morphogenetic protein-independent low-density lipoprotein receptor-related protein 1-extracellular signal-regulated kinase activation, calcineurin signaling, and low-density lipoprotein receptor-related protein 1ß-mediated nuclear factor 45 nuclear export in response to BMPER treatment. CONCLUSIONS: We conclude that BMPER plays a pivotal role in pulmonary inflammatory response, which provides new therapeutic options against sepsis shock. The new signaling pathway initiated by BMPER/low-density lipoprotein receptor-related protein 1 axis broadens our understanding about BMPER's role in vascular homeostasis.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Endotoxinas , Pulmón/irrigación sanguínea , Neumonía/metabolismo , Receptores de LDL/metabolismo , Transducción de Señal , Proteínas Supresoras de Tumor/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Animales , Apoptosis , Permeabilidad Capilar , Proteínas Portadoras/genética , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/patología , Predisposición Genética a la Enfermedad , Haploinsuficiencia , Mediadores de Inflamación/metabolismo , Proteína 1 Relacionada con Receptor de Lipoproteína de Baja Densidad , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción NFATC/metabolismo , Proteína del Factor Nuclear 45/metabolismo , Fenotipo , Neumonía/inducido químicamente , Neumonía/genética , Neumonía/patología , Interferencia de ARN , Receptores de LDL/genética , Factores de Tiempo , Transfección , Proteínas Supresoras de Tumor/genética
12.
EMBO J ; 32(10): 1409-24, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23612611

RESUMEN

Integrin-linked kinase (ILK) is an adaptor protein required to establish and maintain the connection between integrins and the actin cytoskeleton. This linkage is essential for generating force between the extracellular matrix (ECM) and the cell during migration and matrix remodelling. The mechanisms by which ILK stability and turnover are regulated are unknown. Here we report that the E3 ligase CHIP-heat shock protein 90 (Hsp90) axis regulates ILK turnover in fibroblasts. The chaperone Hsp90 stabilizes ILK and facilitates the interaction of ILK with α-parvin. When Hsp90 activity is blocked, ILK is ubiquitinated by CHIP and degraded by the proteasome, resulting in impaired fibroblast migration and a dramatic reduction in the fibrotic response to bleomycin in mice. Together, our results uncover how Hsp90 regulates ILK stability and identify a potential therapeutic strategy to alleviate fibrotic diseases.


Asunto(s)
Movimiento Celular/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Actinas/metabolismo , Animales , Bleomicina/toxicidad , Células Cultivadas , Citoesqueleto/metabolismo , Citoesqueleto/ultraestructura , Matriz Extracelular/metabolismo , Femenino , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Fibrosis/inducido químicamente , Fibrosis/metabolismo , Adhesiones Focales/fisiología , Proteínas HSP90 de Choque Térmico/genética , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Piel/efectos de los fármacos , Piel/patología , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
13.
J Heart Valve Dis ; 26(5): 569-580, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-29762926

RESUMEN

BACKGROUND: A genetic component to familial mitral valve prolapse (MVP) has been proposed for decades. Despite this, very few genes have been linked to MVP. Herein is described a four-generation pedigree with numerous individuals affected with severe MVP, some at strikingly young ages. METHODS: A detailed clinical evaluation performed on all affected family members demonstrated a spectrum of MVP morphologies and associated phenotypes. RESULTS: Linkage analysis failed to identify strong candidate loci, but revealed significant regions, which were investigated further using whole-exome sequencing of one of the severely affected family members. Whole-exome sequencing identified variants in this individual that fell within linkage analysis peak regions, but none was an obvious pathogenic candidate. Follow up segregation analysis of all exome-identified variants was performed to genotype other affected and unaffected individuals in the family, but no variants emerged as clear pathogenic candidates. Two notable variants of uncertain significance in candidate genes were identified: p.I1013S in PTPRJ at 11p11.2 and FLYWCH1 p.R540Q at 16p13.3. Neither gene has been previously linked to MVP in humans, although PTPRJ mutant mice display defects in endocardial cushions, which give rise to the cardiac valves. PTPRJ and FLYWCH1 expression was detected in adult human mitral valve cells, and in-silico analysis of these variants suggests they may be deleterious. However, neither variant segregated completely with all of the affected individuals in the family, particularly when 'affected' was broadly defined. CONCLUSIONS: While a contributory role for PTPRJ and FLYWCH1 in this family cannot be excluded, the study results underscored the difficulties involved in uncovering the genomic contribution to MVP, even in apparently Mendelian families.


Asunto(s)
Prolapso de la Válvula Mitral , Dedos de Zinc/genética , Adulto , Niño , Ecocardiografía/métodos , Salud de la Familia , Femenino , Estudios de Asociación Genética , Humanos , Masculino , Persona de Mediana Edad , Prolapso de la Válvula Mitral/diagnóstico , Prolapso de la Válvula Mitral/genética , Proteínas Tirosina Fosfatasas Clase 3 Similares a Receptores/genética , Secuenciación del Exoma/métodos
14.
J Biol Chem ; 290(14): 9183-94, 2015 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-25691571

RESUMEN

Patients with chronic obstructive pulmonary disease, acute lung injury, and critical care illness may develop hypercapnia. Many of these patients often have muscle dysfunction which increases morbidity and impairs their quality of life. Here, we investigated whether hypercapnia leads to skeletal muscle atrophy. Mice exposed to high CO2 had decreased skeletal muscle wet weight, fiber diameter, and strength. Cultured myotubes exposed to high CO2 had reduced fiber diameter, protein/DNA ratios, and anabolic capacity. High CO2 induced the expression of MuRF1 in vivo and in vitro, whereas MuRF1(-/-) mice exposed to high CO2 did not develop muscle atrophy. AMP-activated kinase (AMPK), a metabolic sensor, was activated in myotubes exposed to high CO2, and loss-of-function studies showed that the AMPKα2 isoform is necessary for muscle-specific ring finger protein 1 (MuRF1) up-regulation and myofiber size reduction. High CO2 induced AMPKα2 activation, triggering the phosphorylation and nuclear translocation of FoxO3a, and leading to an increase in MuRF1 expression and myotube atrophy. Accordingly, we provide evidence that high CO2 activates skeletal muscle atrophy via AMPKα2-FoxO3a-MuRF1, which is of biological and potentially clinical significance in patients with lung diseases and hypercapnia.


Asunto(s)
Adenilato Quinasa/metabolismo , Dióxido de Carbono/metabolismo , Factores de Transcripción Forkhead/metabolismo , Proteínas Musculares/metabolismo , Músculo Esquelético/patología , Atrofia Muscular/etiología , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Secuencia de Bases , Línea Celular , Cartilla de ADN , Proteína Forkhead Box O3 , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas de Motivos Tripartitos , Regulación hacia Arriba
15.
Hum Mol Genet ; 23(4): 1013-24, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24113144

RESUMEN

Gordon Holmes syndrome (GHS) is a rare Mendelian neurodegenerative disorder characterized by ataxia and hypogonadism. Recently, it was suggested that disordered ubiquitination underlies GHS though the discovery of exome mutations in the E3 ligase RNF216 and deubiquitinase OTUD4. We performed exome sequencing in a family with two of three siblings afflicted with ataxia and hypogonadism and identified a homozygous mutation in STUB1 (NM_005861) c.737C→T, p.Thr246Met, a gene that encodes the protein CHIP (C-terminus of HSC70-interacting protein). CHIP plays a central role in regulating protein quality control, in part through its ability to function as an E3 ligase. Loss of CHIP function has long been associated with protein misfolding and aggregation in several genetic mouse models of neurodegenerative disorders; however, a role for CHIP in human neurological disease has yet to be identified. Introduction of the Thr246Met mutation into CHIP results in a loss of ubiquitin ligase activity measured directly using recombinant proteins as well as in cell culture models. Loss of CHIP function in mice resulted in behavioral and reproductive impairments that mimic human ataxia and hypogonadism. We conclude that GHS can be caused by a loss-of-function mutation in CHIP. Our findings further highlight the role of disordered ubiquitination and protein quality control in the pathogenesis of neurodegenerative disease and demonstrate the utility of combining whole-exome sequencing with molecular analyses and animal models to define causal disease polymorphisms.


Asunto(s)
Anomalías Múltiples/enzimología , Ataxia Cerebelosa/enzimología , Hormona Liberadora de Gonadotropina/deficiencia , Hipogonadismo/enzimología , Ubiquitina-Proteína Ligasas/genética , Anomalías Múltiples/genética , Adolescente , Secuencia de Aminoácidos , Animales , Células COS , Ataxia Cerebelosa/genética , Cerebelo/metabolismo , Cerebelo/patología , Chlorocebus aethiops , Femenino , Estudios de Asociación Genética , Hormona Liberadora de Gonadotropina/genética , Humanos , Hipogonadismo/genética , Masculino , Ratones , Datos de Secuencia Molecular , Mutación Missense , Fenotipo , Ubiquitina-Proteína Ligasas/deficiencia , Adulto Joven
16.
Arterioscler Thromb Vasc Biol ; 35(2): 358-67, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25503991

RESUMEN

OBJECTIVE: Previously, we have identified bone morphogenetic protein endothelial cell precursor-derived regulator (BMPER) to increase the angiogenic activity of endothelial cells in a concentration-dependent manner. In this project, we now investigate how BMPER acts in concert with key molecules of angiogenesis to promote blood vessel formation. APPROACH AND RESULTS: To assess the effect of BMPER on angiogenesis-related signaling pathways, we performed an angiogenesis antibody array with BMPER-stimulated endothelial cells. We detected increased basic fibroblast growth factor (bFGF/FGF-2) expression after BMPER stimulation and decreased expression of thrombospondin-1. Additionally, FGF receptor-1 expression, phosphorylation, FGF signaling pathway activity, and cell survival were increased. Consistently, silencing of BMPER by small interfering RNA decreased bFGF and FGF receptor-1 expression and increased thrombospondin-1 expression and cell apoptosis. Next, we investigated the interaction of BMPER and the FGF signaling pathway in endothelial cell function. BMPER stimulation increased endothelial cell angiogenic activity in migration, Matrigel, and spheroid assays. To block FGF signaling, an anti-bFGF antibody was used, which effectively inhibited the proangiogenic BMPER effect. Accordingly, BMPER-silenced endothelial cells under bFGF stimulation showed decreased angiogenic activity compared with bFGF control. We confirmed these findings in vivo by subcutaneous Matrigel injections with and without bFGF in C57BL/6_Bmper(+/-) mice. Aortic ring assays of C57BL/6_Bmper(+/-) mice confirmed a specific effect for bFGF but not for vascular endothelial growth factor. CONCLUSIONS: Taken together, the proangiogenic BMPER effect in endothelial cells is mediated by inhibition of antiangiogenic thrombospondin-1 and enhanced expression and activation of the FGF signaling pathway that is crucial in the promotion of angiogenesis.


Asunto(s)
Proteínas Portadoras/metabolismo , Células Endoteliales/metabolismo , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Neovascularización Fisiológica , Animales , Apoptosis , Proteínas Portadoras/genética , Proteínas Portadoras/farmacología , Células Cultivadas , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 2 de Crecimiento de Fibroblastos/farmacología , Células Endoteliales de la Vena Umbilical Humana/metabolismo , Humanos , Ratones Endogámicos C57BL , Ratones Noqueados , Neovascularización Fisiológica/efectos de los fármacos , Fosforilación , Análisis por Matrices de Proteínas , Proteómica/métodos , Interferencia de ARN , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas Recombinantes/farmacología , Transducción de Señal , Trombospondina 1/metabolismo , Técnicas de Cultivo de Tejidos , Transfección
17.
Mol Cell ; 31(6): 862-72, 2008 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-18922468

RESUMEN

The CRAF protein kinase regulates proliferative, differentiation, and survival signals from activated RAS proteins to downstream effectors, most often by inducing MEK/ERK activation. A well-established model of CRAF regulation involves RAS-mediated translocation of CRAF to the plasma membrane, where it is activated by a series of events including phosphorylation. Here we have discovered a new mode of regulation that occurs prior to this step. By creating a kinase-defective version of CRAF in mice or by use of the RAF inhibitor sorafenib, we show that CRAF must first undergo autophosphorylation of serine 621 (S621). Autophosphorylation occurs in cis, does not involve MEK/ERK activation, and is essential to ensure the correct folding and stability of the protein. In the absence of S621 phosphorylation, CRAF is degraded by the proteasome by mechanisms that do not uniquely rely on the E3 ubiquitin ligase CHIP.


Asunto(s)
Fosfoserina/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas c-raf/metabolismo , Animales , Activación Enzimática , Estabilidad de Enzimas , Fibroblastos/enzimología , Ratones , Modelos Biológicos , Fenotipo , Fosforilación , Pliegue de Proteína , Ubiquitinación
18.
J Mol Cell Cardiol ; 80: 156-65, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25633836

RESUMEN

PHD3, a member of a family of Prolyl-4 Hydroxylase Domain (PHD) proteins, has long been considered a pro-apoptotic protein. Although the pro-apoptotic effect of PHD3 requires its prolyl hydroxylase activity, it may be independent of HIF-1α, the common substrate of PHDs. PHD3 is highly expressed in the heart, however, its role in cardiomyocyte apoptosis remains unclear. This study was undertaken to determine whether inhibition or depletion of PHD3 inhibits cardiomyocyte apoptosis and attenuates myocardial injury induced by ischemia-reperfusion (I/R). PHD3 knockout mice and littermate controls were subjected to left anterior descending (LAD) coronary artery ligation for 40 min followed by reperfusion. Histochemical analysis using Evan's Blue, triphenyl-tetrazolium chloride and TUNEL staining, demonstrated that myocardial injury and cardiomyocyte apoptosis induced I/R injury were significantly attenuated in PHD3 knockout mice. PHD3 knockout mice exhibited no changes in HIF-1α protein level, the expression of some HIF target genes or the myocardium capillary density at physiological condition. However, depletion of PHD3 further enhanced the induction of HIF-1α protein at hypoxic condition and increased expression of HIF-1α inhibited cardiomyocyte apoptosis induced by hypoxia. In addition, it has been demonstrated that PHD3 plays an important role in ATR/Chk1/p53 pathway. Consistently, a prolyl hydroxylase inhibitor or depletion of PHD3 significantly inhibits the activation of Chk1 and p53 in cardiomyocytes and the subsequent apoptosis induced by doxorubicin, hydrogen peroxide or hypoxia/reoxygenation. Taken together, these data suggest that depletion of PHD3 leads to increased stabilization of HIF-1α and inhibition of DNA damage response, both of which may contribute to the cardioprotective effect seen with depletion of PHD3.


Asunto(s)
Apoptosis/genética , Daño por Reperfusión Miocárdica/genética , Miocitos Cardíacos/metabolismo , Procolágeno-Prolina Dioxigenasa/genética , Animales , Animales Recién Nacidos , Apoptosis/efectos de los fármacos , Línea Celular , Daño del ADN/efectos de los fármacos , Modelos Animales de Enfermedad , Doxorrubicina/farmacología , Expresión Génica , Genotipo , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Ratones , Ratones Noqueados , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocitos Cardíacos/efectos de los fármacos , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/genética , Procolágeno-Prolina Dioxigenasa/metabolismo , Ratas
19.
Dev Biol ; 395(1): 111-9, 2014 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-25173872

RESUMEN

The establishment of the coronary circulation is one of the final critical steps during heart development. Despite decades of research, our understanding of how the coronary vasculature develops and connects to the aorta remains limited. This review serves two specific purposes: it addresses recent advances in understanding the origin of the coronary endothelium, and it then focuses on the last crucial step of coronary vasculature development, the connection of the coronary plexus to the aorta. The chick and quail animal models have yielded most of the information for how these connections form, starting with a fine network of vessels that penetrate the aorta and coalesce to form two distinct ostia. Studies in mouse and rat confirm that at least some of these steps are conserved in mammals, but gaps still exist in our understanding of mammalian coronary ostia formation. The signaling cues necessary to guide the coronary plexus to the aorta are also incompletely understood. Hypoxia-inducible transcription factor-1 and its downstream targets are among the few identified genes that promote the formation of the coronary stems. Together, this review summarizes our current knowledge of coronary vascular formation and highlights the significant gaps that remain. In addition, it highlights some of the coronary artery anomalies known to affect human health, demonstrating that even seemingly subtle defects arising from incorrect coronary plexus formation can result in significant health crises.


Asunto(s)
Vasos Coronarios/embriología , Endotelio Vascular/embriología , Corazón/embriología , Modelos Anatómicos , Modelos Cardiovasculares , Animales , Vasos Coronarios/citología , Vasos Coronarios/metabolismo , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Corazón/anatomía & histología , Humanos , Células Madre/citología , Células Madre/metabolismo
20.
Dev Biol ; 386(2): 385-94, 2014 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-24373957

RESUMEN

The connection of the coronary vasculature to the aorta is one of the last essential steps of cardiac development. However, little is known about the signaling events that promote normal coronary artery formation. The bone morphogenetic protein (BMP) signaling pathway regulates multiple aspects of endothelial cell biology but has not been specifically implicated in coronary vascular development. BMP signaling is tightly regulated by numerous factors, including BMP-binding endothelial cell precursor-derived regulator (BMPER), which can both promote and repress BMP signaling activity. In the embryonic heart, BMPER expression is limited to the endothelial cells and the endothelial-derived cushions, suggesting that BMPER may play a role in coronary vascular development. Histological analysis of BMPER(-/-) embryos at early embryonic stages demonstrates that commencement of coronary plexus differentiation is normal and that endothelial apoptosis and cell proliferation are unaffected in BMPER(-/-) embryos compared with wild-type embryos. However, analysis between embryonic days 15.5-17.5 reveals that, in BMPER(-/-) embryos, coronary arteries are either atretic or connected distal to the semilunar valves. In vitro tubulogenesis assays indicate that isolated BMPER(-/-) endothelial cells have impaired tube formation and migratory ability compared with wild-type endothelial cells, suggesting that these defects may lead to the observed coronary artery anomalies seen in BMPER(-/-) embryos. Additionally, recombinant BMPER promotes wild-type ventricular endothelial migration in a dose-dependent manner, with a low concentration promoting and high concentrations inhibiting migration. Together, these results indicate that BMPER-regulated BMP signaling is critical for coronary plexus remodeling and normal coronary artery development.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Proteínas Portadoras/metabolismo , Vasos Coronarios/fisiología , Células Endoteliales/metabolismo , Transducción de Señal/fisiología , Análisis de Varianza , Animales , Proteínas Portadoras/genética , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Ratones , Ratones Noqueados , Microscopía Fluorescente
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA