Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 320(6): H2401-H2415, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33989080

RESUMEN

Cardiovascular adaptation underlies all athletic training modalities, with a variety of factors contributing to overall response during exercise-induced stimulation. In this regard the role of circulating biomarkers is a well-established and invaluable tool for monitoring cardiovascular function. Specifically, novel biomarkers such as circulating cell free DNA and RNA are now becoming attractive tools for monitoring cardiovascular function with the advent of next generation technologies that can provide unprecedented precision and resolution of these molecular signatures, paving the way for novel diagnostic and prognostic avenues to better understand physiological remodeling that occurs in trained versus untrained states. In particular, microRNAs are a species of regulatory RNAs with pleiotropic effects on multiple pathways in tissue-specific manners. Furthermore, the identification of cell free microRNAs within peripheral circulation represents a distal signaling mechanism that is just beginning to be explored via a diversity of molecular and bioinformatic approaches. This article provides an overview of the emerging field of sports/performance genomics with a focus on the role of microRNAs as novel functional diagnostic and prognostic tools, and discusses present knowledge in the context of athletic vascular remodeling. This review concludes with current advantages and limitations, touching upon future directions and implications for applying contemporary systems biology knowledge of exercise-induced physiology to better understand how disruption can lead to pathology.


Asunto(s)
MicroARN Circulante/genética , Endotelio Vascular/metabolismo , Ejercicio Físico/fisiología , Remodelación Vascular/genética , Animales , Ácidos Nucleicos Libres de Células , MicroARN Circulante/metabolismo , Endotelio Vascular/fisiología , Entrenamiento Aeróbico , Humanos , Inflamación/genética , Neovascularización Fisiológica/genética , Condicionamiento Físico Animal/fisiología , Estrés Mecánico , Trombosis/genética , Remodelación Vascular/fisiología , Sistema Vasomotor/metabolismo , Sistema Vasomotor/fisiología
2.
J Mol Cell Cardiol ; 141: 43-52, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32209327

RESUMEN

Cardiovascular disease is a pressing health problem with significant global health, societal, and financial burdens. Understanding the molecular basis of polygenic cardiac pathology is thus essential to devising novel approaches for management and treatment. Recent identification of uncharacterized regulatory functions for a class of nuclear envelope proteins called nucleoporins offers the opportunity to understand novel putative mechanisms of cardiac disease development and progression. Consistent reports of nucleoporin deregulation associated with ischemic and dilated cardiomyopathies, arrhythmias and valvular disorders suggests that nucleoporin impairment may be a significant but understudied variable in cardiopathologic disorders. This review discusses and converges existing literature regarding nuclear pore complex proteins and their association with cardiac pathologies, and proposes a role for nucleoporins as facilitators of cardiac disease.


Asunto(s)
Enfermedades Cardiovasculares/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Animales , Sistema Cardiovascular/crecimiento & desarrollo , Sistema Cardiovascular/metabolismo , Humanos , Modelos Biológicos
3.
Can J Physiol Pharmacol ; 97(4): 257-264, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30388388

RESUMEN

Nucleoporins are a specialized subset of nuclear proteins that comprise the nuclear pore complex and regulate nucleocytoplasmic transport. Recent demonstrations of roles for individual nucleoporins in multiple paradigms of differentiation via mechanisms independent of nuclear trafficking represent conceptual advances in understanding the contributions of nucleoporins to cellular development. Among these, a functional role for nucleoporins in reproductive fitness and gametogenesis has been identified, supported by robust models and clinical studies that leverage the power of next generation sequencing technology to identify reproductive-disease-associated mutations in specific nucleoporins. Proper nucleoporin function manifests in different ways during oogenesis and spermatogenesis. However, nonhuman models of gametogenesis may not recapitulate human mechanisms, which may confound translational interpretation and relevance. To circumvent these limitations, identification of reproductive pathologies in patients, combined with next generation sequencing approaches and advanced in silico tools, offers a powerful approach to investigate the potential function of nucleoporins in human reproduction. Ultimately, elucidating the role of nucleoporins in reproductive biology will provide opportunities for predictive, diagnostic, and therapeutic strategies to address reproductive disorders.


Asunto(s)
Proteínas de Complejo Poro Nuclear/metabolismo , Reproducción , Animales , Crecimiento y Desarrollo , Humanos , Oogénesis , Espermatogénesis , Investigación Biomédica Traslacional
4.
J Mol Cell Cardiol ; 92: 63-74, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26826378

RESUMEN

Calreticulin deficiency causes myocardial developmental defects that culminate in an embryonic lethal phenotype. Recent studies have linked loss of this calcium binding chaperone to failure in myofibrillogenesis through an as yet undefined mechanism. The purpose of the present study was to identify cellular processes corrupted by calreticulin deficiency that precipitate dysregulation of cardiac myofibrillogenesis related to acquisition of cardiac phenotype. In an embryonic stem cell knockout model, calreticulin deficit (crt(-/-)) compromised nucleocytoplasmic transport of nuclear localization signal-dependent and independent pathways, disrupting nuclear import of the cardiac transcription factor MEF2C. The expression of nucleoporins and associated nuclear transport proteins in derived crt(-/-) cardiomyocytes revealed an abnormal nuclear pore complex (NPC) configuration. Altered protein content in crt(-/-) cells resulted in remodeled NPC architecture that caused decreased pore diameter and diminished probability of central channel occupancy versus wild type counterparts. Ionophore treatment of impaired calcium handling in crt(-/-) cells corrected nuclear pore microarchitecture and rescued nuclear import resulting in normalized myofibrillogenesis. Thus, calreticulin deficiency alters nuclear pore function and structure, impeding myofibrillogenesis in nascent cardiomyocytes through a calcium dependent mechanism. This essential role of calreticulin in nucleocytoplasmic communication competency ties its regulatory action with proficiency of cardiac myofibrillogenesis essential for proper cardiac development.


Asunto(s)
Calreticulina/genética , Cardiomiopatías/genética , Desarrollo de Músculos/genética , Poro Nuclear/genética , Transporte Activo de Núcleo Celular/genética , Animales , Calcio/metabolismo , Señalización del Calcio/genética , Calreticulina/deficiencia , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Células Madre Embrionarias/patología , Técnicas de Inactivación de Genes , Humanos , Factores de Transcripción MEF2/genética , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Miocitos Cardíacos/ultraestructura , Poro Nuclear/metabolismo , Poro Nuclear/ultraestructura
5.
Am J Physiol Heart Circ Physiol ; 321(4): H665-H666, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34524924
6.
J Exp Med ; 204(2): 405-20, 2007 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-17283208

RESUMEN

Embryonic stem cells have the distinct potential for tissue regeneration, including cardiac repair. Their propensity for multilineage differentiation carries, however, the liability of neoplastic growth, impeding therapeutic application. Here, the tumorigenic threat associated with embryonic stem cell transplantation was suppressed by cardiac-restricted transgenic expression of the reprogramming cytokine TNF-alpha, enhancing the cardiogenic competence of recipient heart. The in vivo aptitude of TNF-alpha to promote cardiac differentiation was recapitulated in embryoid bodies in vitro. The procardiogenic action required an intact endoderm and was mediated by secreted cardio-inductive signals. Resolved TNF-alpha-induced endoderm-derived factors, combined in a cocktail, secured guided differentiation of embryonic stem cells in monolayers produce cardiac progenitors termed cardiopoietic cells. Characterized by a down-regulation of oncogenic markers, up-regulation, and nuclear translocation of cardiac transcription factors, this predetermined population yielded functional cardiomyocyte progeny. Recruited cardiopoietic cells delivered in infarcted hearts generated cardiomyocytes that proliferated into scar tissue, integrating with host myocardium for tumor-free repair. Thus, cardiopoietic programming establishes a strategy to hone stem cell pluripotency, offering a tumor-resistant approach for regeneration.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Corazón/fisiología , Miocitos Cardíacos/citología , Regeneración/fisiología , Trasplante de Células Madre/métodos , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Cartilla de ADN , Células Madre Embrionarias/trasplante , Regulación de la Expresión Génica , Ratones , Ratones Transgénicos , Análisis por Micromatrices , Neoplasias/prevención & control , Factores de Transcripción/metabolismo
7.
Croat Med J ; 54(4): 319-29, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23986272

RESUMEN

Development of innovative high throughput technologies has enabled a variety of molecular landscapes to be interrogated with an unprecedented degree of detail. Emergence of next generation nucleotide sequencing methods, advanced proteomic techniques, and metabolic profiling approaches continue to produce a wealth of biological data that captures molecular frameworks underlying phenotype. The advent of these novel technologies has significant translational applications, as investigators can now explore molecular underpinnings of developmental states with a high degree of resolution. Application of these leading-edge techniques to patient samples has been successfully used to unmask nuanced molecular details of disease vs healthy tissue, which may provide novel targets for palliative intervention. To enhance such approaches, concomitant development of algorithms to reprogram differentiated cells in order to recapitulate pluripotent capacity offers a distinct advantage to advancing diagnostic methodology. Bioinformatic deconvolution of several "-omic" layers extracted from reprogrammed patient cells, could, in principle, provide a means by which the evolution of individual pathology can be developmentally monitored. Significant logistic challenges face current implementation of this novel paradigm of patient treatment and care, however, several of these limitations have been successfully addressed through continuous development of cutting edge in silico archiving and processing methods. Comprehensive elucidation of genomic, transcriptomic, proteomic, and metabolomic networks that define normal and pathological states, in combination with reprogrammed patient cells are thus poised to become high value resources in modern diagnosis and prognosis of patient disease.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Técnicas de Diagnóstico Molecular , Sistemas de Atención de Punto , Proteómica/métodos , Trasplante de Células Madre , Atención a la Salud/métodos , Humanos
8.
Results Probl Cell Differ ; 70: 581-593, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36348122

RESUMEN

The nucleus is a complex organelle with functions beyond being a simple repository for genomic material. For example, its actions in biomechanical sensing, protein synthesis, and epigenomic regulation showcase how the nucleus integrates multiple signaling modalities to intricately regulate gene expression. This innate dynamism is underscored by subnuclear components that facilitate these roles, with elements of the nucleoskeleton, phase-separated nuclear bodies, and chromatin safeguarding by nuclear envelope proteins providing examples of this functional diversity. Among these, one of the lesser characterized nuclear organelles is the nucleolar channel system (NCS), first reported several decades ago in human endometrial biopsies. This tubular structure, believed to be derived from the inner nuclear membrane of the nuclear envelope, was first observed in secretory endometrial cells during a specific phase of the menstrual cycle. Reported as a consistent, yet transient, nuclear organelle, current interpretations of existing data suggest that it serves as a marker of a window for optimal implantation. In spite of this available data, the NCS remains incompletely characterized structurally and functionally, due in part to its transient spatial and temporal expression. As a further complication, evidence exists showing NCS expression in fetal tissue, suggesting that it may not act exclusively as a marker of uterine receptivity, but rather as a hormone sensor sensitive to estrogen and progesterone ratios. To gain a better understanding of the NCS, current technologies can be applied to profile rare cell populations or capture transient structural dynamics, for example, at a level of sensitivity and resolution not previously possible. Moving forward, advanced characterization of the NCS will shed light on an uncharacterized aspect of reproductive physiology, with the potential to refine assisted reproductive techniques.


Asunto(s)
Implantación del Embrión , Endometrio , Femenino , Humanos , Endometrio/metabolismo , Técnicas Reproductivas Asistidas , Núcleo Celular
9.
J Cell Mol Med ; 15(4): 928-37, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20497490

RESUMEN

Mitogen activated protein (MAP) kinases control eukaryotic proliferation, and import of kinases into the nucleus through the nuclear pore complex (NPC) can influence gene expression to affect cellular growth, cell viability and homeostatic function. The NPC is a critical regulatory checkpoint for nucleocytoplasmic traffic that regulates gene expression and cell growth, and MAP kinases may be physically associated with the NPC to modulate transport. In the present study, highly enriched NPC fractions were isolated and investigated for associated kinases and/or activity. Endogenous kinase activity was identified within the NPC fraction, which phosphorylated a 30 kD nuclear pore protein. Phosphomodification of this nucleoporin, here termed Nup30, was inhibited by apigenin and PD-98059, two MAP kinase antagonists as well as with SB-202190, a pharmacological blocker of p38. Furthermore, high throughput profiling of enriched NPCs revealed constitutive presence of all members of the MAP kinase family, extracellular regulated kinases (ERK), p38 and Jun N-terminal kinase. The NPC thus contains a spectrum of associated MAP kinases that suggests an intimate role for ERK and p38 in regulation of nuclear pore function.


Asunto(s)
Proteínas Quinasas Activadas por Mitógenos/metabolismo , Poro Nuclear/enzimología , Animales , Calcio/metabolismo , Proliferación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/enzimología , Poro Nuclear/efectos de los fármacos , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Ratas , Ratas Sprague-Dawley , Fracciones Subcelulares/efectos de los fármacos , Fracciones Subcelulares/enzimología , Especificidad por Sustrato/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
10.
Stem Cells ; 28(7): 1281-91, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20506533

RESUMEN

Genomic perturbations that challenge normal signaling at the pluripotent stage may trigger unforeseen ontogenic aberrancies. Anticipatory systems biology identification of transcriptome landscapes that underlie latent phenotypes would offer molecular diagnosis before the onset of symptoms. The purpose of this study was to assess the impact of calreticulin-deficient embryonic stem cell transcriptomes on molecular functions and physiological systems. Bioinformatic surveillance of calreticulin-null stem cells, a monogenic insult model, diagnosed a disruption in transcriptome dynamics, which re-prioritized essential cellular functions. Calreticulin-calibrated signaling axes were uncovered, and network-wide cartography of undifferentiated stem cell transcripts suggested cardiac manifestations. Calreticulin-deficient stem cell-derived cardiac cells verified disorganized sarcomerogenesis, mitochondrial paucity, and cytoarchitectural aberrations to validate calreticulin-dependent network forecasts. Furthermore, magnetic resonance imaging and histopathology detected a ventricular septal defect, revealing organogenic manifestation of calreticulin deletion. Thus, bioinformatic deciphering of a primordial calreticulin-deficient transcriptome decoded at the pluripotent stem cell stage a reconfigured multifunctional molecular registry to anticipate predifferentiation susceptibility toward abnormal cardiophenotype.


Asunto(s)
Calreticulina/metabolismo , Células Madre Embrionarias/metabolismo , Miocardio/metabolismo , Animales , Calreticulina/deficiencia , Muerte Celular , Diferenciación Celular , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Células Madre Embrionarias/citología , Perfilación de la Expresión Génica , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Miocardio/citología , Fenotipo
11.
J Am Heart Assoc ; 10(16): e019948, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34369184

RESUMEN

Background Age-related heart diseases are significant contributors to increased morbidity and mortality. Emerging evidence indicates that mitochondria within cardiomyocytes contribute to age-related increased reactive oxygen species (ROS) generation that plays an essential role in aging-associated cardiac diseases. Methods and Results The present study investigated differences between ROS production in cardiomyocytes isolated from adult (6 months) and aged (24 months) Fischer 344 rats, and in cardiac tissue of adult (18-65 years) and elderly (>65 years) patients with preserved cardiac function. Superoxide dismutase inhibitable ferricytochrome c reduction assay (1.32±0.63 versus 0.76±0.31 nMol/mg per minute; P=0.001) superoxide and H2O2 production, measured as dichlorofluorescein diacetate fluorescence (1646±428 versus 699±329, P=0.04), were significantly higher in the aged versus adult cardiomyocytes. Similarity in age-related alteration between rats and humans was identified in mitochondrial-electron transport chain-complex-I-associated increased oxidative-stress by MitoSOX fluorescence (53.66±18.58 versus 22.81±12.60; P=0.03) and in 4-HNE adduct levels (187.54±54.8 versus 47.83±16.7 ng/mg protein, P=0.0063), indicative of increased peroxidation in the elderly. These differences correlated with changes in functional enrichment of genes regulating ROS homeostasis pathways in aged human and rat hearts. Functional merged collective network and pathway enrichment analysis revealed common genes prioritized in human and rat aging-associated networks that underlay enriched functional terms of mitochondrial complex I and common pathways in the aging human and rat heart. Conclusions Aging sensitizes mitochondrial and extramitochondrial mechanisms of ROS buildup within the heart. Network analysis of the transcriptome highlights the critical elements involved with aging-related ROS homeostasis pathways common in rat and human hearts as targets.


Asunto(s)
Envejecimiento/metabolismo , Metabolismo Energético , Mitocondrias Cardíacas/metabolismo , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , Especies Reactivas de Oxígeno/metabolismo , Transcripción Genética , Transcriptoma , Adolescente , Adulto , Factores de Edad , Anciano , Envejecimiento/genética , Animales , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Metabolismo Energético/genética , Femenino , Redes Reguladoras de Genes , Humanos , Peroxidación de Lípido , Masculino , Persona de Mediana Edad , Mitocondrias Cardíacas/genética , Fosforilación Oxidativa , Estrés Oxidativo/genética , Ratas Endogámicas F344 , Superóxido Dismutasa/genética , Superóxido Dismutasa/metabolismo , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Adulto Joven
12.
J Mol Cell Cardiol ; 48(4): 725-34, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20045004

RESUMEN

Decoding of the bioenergetic signature underlying embryonic stem cell cardiac differentiation has revealed a mandatory transformation of the metabolic infrastructure with prominent mitochondrial network expansion and a distinctive switch from glycolysis to oxidative phosphorylation. Here, we demonstrate that despite reduction in total glycolytic capacity, stem cell cardiogenesis engages a significant transcriptome, proteome, as well as enzymatic and topological rearrangement in the proximal, medial, and distal modules of the glycolytic pathway. Glycolytic restructuring was manifested by a shift in hexokinase (Hk) isoforms from Hk-2 to cardiac Hk-1, with intracellular and intermyofibrillar localization mapping mitochondrial network arrangement. Moreover, upregulation of cardiac-specific enolase 3, phosphofructokinase, and phosphoglucomutase and a marked increase in glyceraldehyde 3-phosphate dehydrogenase (GAPDH) phosphotransfer activity, along with apparent post-translational modifications of GAPDH and phosphoglycerate kinase, were all distinctive for derived cardiomyocytes compared to the embryonic stem cell source. Lactate dehydrogenase (LDH) isoforms evolved towards LDH-2 and LDH-3, containing higher proportions of heart-specific subunits, and pyruvate dehydrogenase isoforms rearranged between E1alpha and E1beta, transitions favorable for substrate oxidation in mitochondria. Concomitantly, transcript levels of fetal pyruvate kinase isoform M2, aldolase 3, and transketolase, which shunt the glycolytic with pentose phosphate pathways, were reduced. Collectively, changes in glycolytic pathway modules indicate active redeployment, which would facilitate connectivity of the expanding mitochondrial network with ATP utilization sites. Thus, the delineated developmental dynamics of the glycolytic phosphotransfer network is integral to the remodeling of cellular energetic infrastructure underlying stem cell cardiogenesis.


Asunto(s)
Células Madre Embrionarias/citología , Animales , Diferenciación Celular , Electroforesis en Gel Bidimensional , Perfilación de la Expresión Génica , Gliceraldehído-3-Fosfato Deshidrogenasas/metabolismo , Glucólisis , Procesamiento de Imagen Asistido por Computador , L-Lactato Deshidrogenasa/metabolismo , Metabolómica , Ratones , Vía de Pentosa Fosfato , Fosfoglicerato Quinasa/metabolismo , Isoformas de Proteínas , Procesamiento Proteico-Postraduccional
13.
Differentiation ; 77(3): 248-55, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19272523

RESUMEN

Embryonic stem cell differentiation recapitulates the diverse phenotypes of a developing embryo, traceable according to markers of lineage specification. At gastrulation, the vascular endothelial growth factor (VEGF) receptor, Flk-1 (KDR), identifies a mesoderm-restricted potential of embryonic stem cells. The multi-lineage propensity of Flk-1(+) progenitors mandates the mapping of fate-modifying co-factors in order to stratify differentiating cytotypes and predict lineage competency. Here, Flk-1-based selection of early embryonic stem cell progeny separated a population depleted of pluripotent (Oct4, Sox2) and endoderm (Sox17) markers. The gene expression profile of the Flk-1(+) population was notable for a significant upregulation in the vasculogenic Sox7 transcription factor, which overlapped with the emergence of primordial cardiac transcription factors GATA-4, Myocardin and Nkx2.5. Sorting the parental Flk-1(+) pool with the chemokine receptor CXCR4 to enrich the cardiopoietic subpopulation uncovered divergent Sox7 expression, with a 7-fold induction in non-cardiac compared to cardiac progenitors. Bioinformatic resolution sequestered a framework of gene expression relationship between Sox transcription factor family members and the Flk-1/CXCR4 axes with significant integration of beta-catenin signaling. Thus, differential Sox7 gene expression presents a novel biomarker profile, and possible regulatory switch, to distinguish cardiovascular pedigrees within Flk-1(+) multi-lineage progenitors.


Asunto(s)
Diferenciación Celular , Linaje de la Célula , Células Madre Embrionarias/citología , Regulación del Desarrollo de la Expresión Génica , Miocitos Cardíacos/citología , Factores de Transcripción SOXF/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Animales , Línea Celular , Perfilación de la Expresión Génica , Ratones , Factores de Transcripción SOXF/clasificación , Transducción de Señal
15.
Artículo en Inglés | MEDLINE | ID: mdl-32118046

RESUMEN

Functional variants in nuclear envelope genes are implicated as underlying causes of cardiopathology. To examine the potential association of single nucleotide variants of nucleoporin genes with cardiac disease, we employed a prognostic scoring approach to investigate variants of NUP155, a nucleoporin gene clinically linked with atrial fibrillation. Here we implemented bioinformatic profiling and predictive scoring, based on the gnomAD, National Heart Lung and Blood Institute-Exome Sequencing Project (NHLBI-ESP) Exome Variant Server, and dbNSFP databases to identify rare single nucleotide variants (SNVs) of NUP155 potentially associated with cardiopathology. This predictive scoring revealed 24 SNVs of NUP155 as potentially cardiopathogenic variants located primarily in the N-terminal crescent-shaped domain of NUP155. In addition, a predicted NUP155 R672G variant prioritized in our study was mapped to a region within the alpha helical stack of the crescent domain of NUP155. Bioinformatic analysis of inferred protein-protein interactions of NUP155 revealed over representation of top functions related to molecular transport, RNA trafficking, and RNA post-transcriptional modification. Topology analysis revealed prioritized hubs critical for maintaining network integrity and informational flow that included FN1, SIRT7, and CUL7 with nodal enrichment of RNA helicases in the topmost enriched subnetwork. Furthermore, integration of the top 5 subnetworks to capture network topology of an expanded framework revealed that FN1 maintained its hub status, with elevation of EED, CUL3, and EFTUD2. This is the first study to report novel discovery of a NUP155 subdomain hotspot that enriches for allelic variants of NUP155 predicted to be clinically damaging, and supports a role for RNA metabolism in cardiac disease and development.

16.
Front Endocrinol (Lausanne) ; 11: 570846, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33042024

RESUMEN

Background: Children born to diabetic or obese mothers have a higher risk of heart disease at birth and later in life. Using chromatin immunoprecipitation sequencing, we previously demonstrated that late-gestation diabetes, maternal high fat (HF) diet, and the combination causes distinct fuel-mediated epigenetic reprogramming of rat cardiac tissue during fetal cardiogenesis. The objective of the present study was to investigate the overall transcriptional signature of newborn offspring exposed to maternal diabetes and maternal H diet. Methods: Microarray gene expression profiling of hearts from diabetes exposed, HF diet exposed, and combination exposed newborn rats was compared to controls. Functional annotation, pathway and network analysis of differentially expressed genes were performed in combination exposed and control newborn rat hearts. Further downstream metabolic assessments included measurement of total and phosphorylated AKT2 and GSK3ß, as well as quantification of glycolytic capacity by extracellular flux analysis and glycogen staining. Results: Transcriptional analysis identified significant fuel-mediated changes in offspring cardiac gene expression. Specifically, functional pathways analysis identified two key signaling cascades that were functionally prioritized in combination exposed offspring hearts: (1) downregulation of fibroblast growth factor (FGF) activated PI3K/AKT pathway and (2) upregulation of peroxisome proliferator-activated receptor gamma coactivator alpha (PGC1α) mitochondrial biogenesis signaling. Functional metabolic and histochemical assays supported these transcriptome changes, corroborating diabetes- and diet-induced cardiac transcriptome remodeling and cardiac metabolism in offspring. Conclusion: This study provides the first data accounting for the compounding effects of maternal hyperglycemia and hyperlipidemia on the developmental cardiac transcriptome, and elucidates nuanced and novel features of maternal diabetes and diet on regulation of heart health.


Asunto(s)
Diabetes Gestacional/metabolismo , Dieta Alta en Grasa/efectos adversos , Redes Reguladoras de Genes/fisiología , Fenómenos Fisiologicos Nutricionales Maternos/fisiología , Miocardio/metabolismo , Transcriptoma/fisiología , Animales , Animales Recién Nacidos , Diabetes Gestacional/genética , Diabetes Gestacional/patología , Dieta Alta en Grasa/tendencias , Femenino , Perfilación de la Expresión Génica/métodos , Masculino , Miocardio/patología , Biogénesis de Organelos , Embarazo , Ratas
17.
Stem Cells ; 26(2): 387-400, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17991915

RESUMEN

In the developing embryo, instructive guidance from the ventral endoderm secures cardiac program induction within the anterolateral mesoderm. Endoderm-guided cardiogenesis, however, has yet to be resolved at the proteome level. Here, through cardiopoietic priming of the endoderm with the reprogramming cytokine tumor necrosis factor alpha (TNFalpha), candidate effectors of embryonic stem cell cardiac differentiation were delineated by comparative proteomics. Differential two-dimensional gel electrophoretic mapping revealed that more than 75% of protein species increased >1.5-fold in the TNFalpha-primed versus unprimed endodermal secretome. Protein spot identification by linear ion trap quadrupole (LTQ) tandem mass spectrometry (MS/MS) and validation by shotgun LTQ-Fourier transform MS/MS following multidimensional chromatography mapped 99 unique proteins from 153 spot assignments. A definitive set of 48 secretome proteins was deduced by iterative bioinformatic screening using algorithms for detection of canonical and noncanonical indices of secretion. Protein-protein interaction analysis, in conjunction with respective expression level changes, revealed a nonstochastic TNFalpha-centric secretome network with a scale-free hierarchical architecture. Cardiovascular development was the primary developmental function of the resolved TNFalpha-anchored network. Functional cooperativity of the derived cardioinductive network was validated through direct application of the TNFalpha-primed secretome on embryonic stem cells, potentiating cardiac commitment and sarcomerogenesis. Conversely, inhibition of primary network hubs negated the procardiogenic effects of TNFalpha priming. Thus, proteomic cartography establishes a systems biology framework for the endodermal secretome network guiding stem cell cardiopoiesis.


Asunto(s)
Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Corazón Fetal/citología , Factor de Necrosis Tumoral alfa/farmacología , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/citología , Endodermo/citología , Endodermo/efectos de los fármacos , Endodermo/metabolismo , Corazón Fetal/efectos de los fármacos , Corazón Fetal/embriología , Corazón Fetal/metabolismo , Ratones , Mapeo de Interacción de Proteínas , Proteoma/aislamiento & purificación , Proteómica , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masas en Tándem
18.
Stem Cells ; 26(6): 1464-73, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18369102

RESUMEN

Pluripotent stem cells demonstrate an inherent propensity for unrestricted multi-lineage differentiation. Translation into regenerative applications requires identification and isolation of tissue-specified progenitor cells. From a comprehensive pool of 11,272 quality-filtered genes, profiling embryonic stem cells at discrete stages of cardiopoiesis revealed 736 transcripts encoding membrane-associated proteins, where 306 were specifically upregulated with cardiogenic differentiation. Bioinformatic dissection of exposed surface biomarkers prioritized the chemokine receptor cluster as the most significantly over-represented gene receptor family during pre cardiac induction, with CXCR4 uniquely associated with mesendoderm formation. CXCR4(+) progenitors were sorted from the embryonic stem cell pool into mesoderm-restricted progeny according to co-expression with the early mesoderm marker Flk-1. In contrast to CXCR4(-)/Flk-1(-) cells, the CXCR4(+)/Flk-1(+) subpopulation demonstrated overexpressed cardiac lineage transcription factors (Mef2C, Myocardin, Nkx2.5), whereas pluripotent genes (Oct4, Fgf4, Sox2) as well as neuroectoderm (Sox1) and endoderm alpha-fetoprotein markers were all depleted. In fact, the CXCR4(+)/Flk-1(+) biomarker combination identified embryonic stem cell progeny significantly enriched with Mesp-1, GATA-4, and Tbx5, indicative of pre cardiac mesoderm and the primary heart field. Although the CXCR4(+)/Flk-1(+) transcriptome shared 97% identity with the CXCR4(-)/Flk-1(-) counterpart, the 818 divergent gene set represented predominantly cardiovascular developmental functions and formed a primitive cardiac network. Differentiation of CXCR4(+)/Flk-1(+) progenitors yielded nuclear translocation of myocardial transcription factors and robust sarcomerogenesis with nascent cardiac tissue demonstrating beating activity and calcium transients. Thus, the CXCR4/Flk-1 biomarker pair predicts the emergence of cardiogenic specification within a pluripotent stem cell pool, enabling targeted selection of cardiopoietic lineage. Disclosure of potential conflicts of interest is found at the end of this article.


Asunto(s)
Células Madre Embrionarias/citología , Miocitos Cardíacos/fisiología , Receptores CXCR4/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Biomarcadores/análisis , Calcio/fisiología , Diferenciación Celular , ADN Complementario/genética , Células Madre Embrionarias/fisiología , Endodermo/citología , Endodermo/fisiología , Perfilación de la Expresión Génica , Mesodermo/citología , Mesodermo/fisiología , Ratones , Miocitos Cardíacos/citología , ARN/genética , ARN/aislamiento & purificación
19.
Sci Rep ; 9(1): 12691, 2019 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-31481660

RESUMEN

Nucleoporins have been reported to regulate pluripotent biology, but how they do so remains partially characterized. This study examined the effects of nup155 gene disruption on mouse embryonic stem cells to gain insights into possible mechanisms by which nucleoporins regulate pluripotency in a pro-arrhythmogenic stem cell line. Embryonic stem cells with gene-trapped nup155 exhibited aberrant colony morphology underscored by abnormal transcriptome remodeling. Bioinformatic analysis of whole transcriptome data from nup155+/- embryonic stem cells revealed changes in a variety of non-coding RNA elements, with significant under expression of miR291a, miR291b, miR293, and miR294. These miRNAs are members of the larger regulatory miR290-295 cluster that regulates pluripotency and are controlled by the canonical stem cell-related factors SOX2, OCT4, and NANOG. Expression analysis of these factors revealed downregulation in all three, supported by biochemical profiling and image analysis. These data implicate disruption of the miR-SOX2/OCT4/NANOG regulatory circuit occurs downstream of nup155 gene lesion.


Asunto(s)
Células Madre Embrionarias/metabolismo , Proteínas de Complejo Poro Nuclear/genética , Alelos , Animales , Línea Celular , Regulación hacia Abajo , Células Madre Embrionarias/citología , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Proteínas de Complejo Poro Nuclear/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN no Traducido/metabolismo , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transcriptoma
20.
Nat Commun ; 10(1): 1929, 2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31028265

RESUMEN

Genetically modified mice have advanced our understanding of valve development and disease. Yet, human pathophysiological valvulogenesis remains poorly understood. Here we report that, by combining single cell sequencing and in vivo approaches, a population of human pre-valvular endocardial cells (HPVCs) can be derived from pluripotent stem cells. HPVCs express gene patterns conforming to the E9.0 mouse atrio-ventricular canal (AVC) endocardium signature. HPVCs treated with BMP2, cultured on mouse AVC cushions, or transplanted into the AVC of embryonic mouse hearts, undergo endothelial-to-mesenchymal transition and express markers of valve interstitial cells of different valvular layers, demonstrating cell specificity. Extending this model to patient-specific induced pluripotent stem cells recapitulates features of mitral valve prolapse and identified dysregulation of the SHH pathway. Concurrently increased ECM secretion can be rescued by SHH inhibition, thus providing a putative therapeutic target. In summary, we report a human cell model of valvulogenesis that faithfully recapitulates valve disease in a dish.


Asunto(s)
Células Endoteliales/patología , Proteínas Hedgehog/genética , Prolapso de la Válvula Mitral/patología , Válvula Mitral/patología , Células Madre Pluripotentes/patología , Animales , Antígenos CD/genética , Antígenos CD/metabolismo , Biomarcadores/metabolismo , Proteína Morfogenética Ósea 2/farmacología , Proteínas Relacionadas con las Cadherinas , Cadherinas/genética , Cadherinas/metabolismo , Diferenciación Celular/efectos de los fármacos , Embrión de Mamíferos , Endocardio/metabolismo , Endocardio/patología , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/trasplante , Transición Epitelial-Mesenquimal/efectos de los fármacos , Factor de Transcripción GATA5/genética , Factor de Transcripción GATA5/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Atrios Cardíacos/metabolismo , Atrios Cardíacos/patología , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Válvula Mitral/metabolismo , Prolapso de la Válvula Mitral/genética , Prolapso de la Válvula Mitral/metabolismo , Prolapso de la Válvula Mitral/terapia , Modelos Biológicos , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/metabolismo , Cultivo Primario de Células , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Proteína Wnt3A/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA