Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Heart Circ Physiol ; 326(2): H370-H384, 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38063811

RESUMEN

To identify how cardiomyocyte mechanosensitive signaling pathways are regulated by anisotropic stretch, micropatterned mouse neonatal cardiomyocytes were stretched primarily longitudinally or transversely to the myofiber axis. Four hours of static, longitudinal stretch induced differential expression of 557 genes, compared with 30 induced by transverse stretch, measured using RNA-seq. A logic-based ordinary differential equation model of the cardiac myocyte mechanosignaling network, extended to include the transcriptional regulation and expression of 784 genes, correctly predicted measured expression changes due to anisotropic stretch with 69% accuracy. The model also predicted published transcriptional responses to mechanical load in vitro or in vivo with 63-91% accuracy. The observed differences between transverse and longitudinal stretch responses were not explained by differential activation of specific pathways but rather by an approximately twofold greater sensitivity to longitudinal stretch than transverse stretch. In vitro experiments confirmed model predictions that stretch-induced gene expression is more sensitive to angiotensin II and endothelin-1, via RhoA and MAP kinases, than to the three membrane ion channels upstream of calcium signaling in the network. Quantitative cardiomyocyte gene expression differs substantially with the axis of maximum principal stretch relative to the myofilament axis, but this difference is due primarily to differences in stretch sensitivity rather than to selective activation of mechanosignaling pathways.NEW & NOTEWORTHY Anisotropic stretch applied to micropatterned neonatal mouse ventricular myocytes induced markedly greater acute transcriptional responses when the major axis of stretch was parallel to the myofilament axis than when it was transverse. Analysis with a novel quantitative network model of mechanoregulated cardiomyocyte gene expression suggests that this difference is explained by higher cell sensitivity to longitudinal loading than transverse loading than by the activation of differential signaling pathways.


Asunto(s)
Miocitos Cardíacos , Transducción de Señal , Animales , Ratones , Miocitos Cardíacos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Angiotensina II/farmacología , Regulación de la Expresión Génica , Células Cultivadas , Estrés Mecánico
2.
EMBO J ; 38(2)2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30523147

RESUMEN

Proper temporal and spatial activation of stem cells relies on highly coordinated cell signaling. The primary cilium is the sensory organelle that is responsible for transmitting extracellular signals into a cell. Primary cilium size, architecture, and assembly-disassembly dynamics are under rigid cell cycle-dependent control. Using mouse incisor tooth epithelia as a model, we show that ciliary dynamics in stem cells require the proper functions of a cholesterol-binding membrane glycoprotein, Prominin-1 (Prom1/CD133), which controls sequential recruitment of ciliary membrane components, histone deacetylase, and transcription factors. Nuclear translocation of Prom1 and these molecules is particularly evident in transit amplifying cells, the immediate derivatives of stem cells. The absence of Prom1 impairs ciliary dynamics and abolishes the growth stimulation effects of sonic hedgehog (SHH) treatment, resulting in the disruption of stem cell quiescence maintenance and activation. We propose that Prom1 is a key regulator ensuring appropriate response of stem cells to extracellular signals, with important implications for development, regeneration, and diseases.


Asunto(s)
Antígeno AC133/metabolismo , Cilios/metabolismo , Incisivo/citología , Antígeno AC133/genética , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Humanos , Incisivo/metabolismo , Ratones , Modelos Biológicos , Mutagénesis Sitio-Dirigida , Transporte de Proteínas , Transducción de Señal , Células Madre/citología , Células Madre/metabolismo
3.
Physiol Genomics ; 52(10): 468-477, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32866086

RESUMEN

Much of our understanding of the regulatory mechanisms governing the cell cycle in mammals has relied heavily on methods that measure the aggregate state of a population of cells. While instrumental in shaping our current understanding of cell proliferation, these approaches mask the genetic signatures of rare subpopulations such as quiescent (G0) and very slowly dividing (SD) cells. Results described in this study and those of others using single-cell analysis reveal that even in clonally derived immortalized cancer cells, ∼1-5% of cells can exhibit G0 and SD phenotypes. Therefore to enable the study of these rare cell phenotypes we established an integrated molecular, computational, and imaging approach to track, isolate, and genetically perturb single cells as they proliferate. A genetically encoded cell-cycle reporter (K67p-FUCCI) was used to track single cells as they traversed the cell cycle. A set of R-scripts were written to quantify K67p-FUCCI over time. To enable the further study G0 and SD phenotypes, we retrofitted a live cell imaging system with a micromanipulator to enable single-cell targeting for functional validation studies. Single-cell analysis revealed HT1080 and MCF7 cells had a doubling time of ∼24 and ∼48 h, respectively, with high duration variability in G1 and G2 phases. Direct single-cell microinjection of mRNA encoding (GFP) achieves detectable GFP fluorescence within ∼5 h in both cell types. These findings coupled with the possibility of targeting several hundreds of single cells improves throughput and sensitivity over conventional methods to study rare cell subpopulations.


Asunto(s)
Ciclo Celular/genética , Genes Reporteros , Antígeno Ki-67/genética , Plásmidos/genética , Análisis de la Célula Individual/métodos , Animales , Proliferación Celular/genética , Células Epiteliales/metabolismo , Colorantes Fluorescentes/metabolismo , Vectores Genéticos , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Antígeno Ki-67/metabolismo , Células MCF-7 , Ratones , Microinyecciones , Fenotipo , ARN Mensajero/administración & dosificación , ARN Mensajero/genética , Transducción Genética
4.
J Physiol ; 595(17): 5797-5813, 2017 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-28688178

RESUMEN

KEY POINTS: Changes in gene expression that occur within hours of exposure to hypoxia in in vivo skeletal muscles remain unexplored. Two hours of hypoxia caused significant down-regulation of extracellular matrix genes followed by a shift at 6 h to altered expression of genes associated with the nuclear lumen while respiratory and blood gases were stabilized. Enrichment analysis of mRNAs classified by stability rates suggests an attenuation of post-transcriptional regulation within hours of hypoxic exposure, where PI3K-Akt signalling was suggested to have a nodal role by pathway analysis. Experimental measurements and bioinformatic analyses suggested that the dephosphorylation of Akt after 2 h of hypoxic exposure might deactivate RNA-binding protein BRF1, hence resulting in the selective degradation of mRNAs. ABSTRACT: The effects of acute hypoxia have been widely studied, but there are few studies of transcriptional responses to hours of hypoxia in vivo, especially in hypoxia-tolerant tissues like skeletal muscles. We used RNA-seq to analyse gene expression in plantaris muscles while monitoring respiration, arterial blood gases, and blood glucose in mice exposed to 8% O2 for 2 or 6 h. Rapid decreases in blood gases and a slower reduction in blood glucose suggest stress, which was accompanied by widespread changes in gene expression. Early down-regulation of genes associated with the extracellular matrix was followed by a shift to genes associated with the nuclear lumen. Most of the early down-regulated genes had mRNA half-lives longer than 2 h, suggesting a role for post-transcriptional regulation. These transcriptional changes were enriched in signalling pathways in which the PI3K-Akt signalling pathway was identified as a hub. Our analyses indicated that gene targets of PI3K-Akt but not HIF were enriched in early transcriptional responses to hypoxia. Among the PI3K-Akt targets, 75% could be explained by a deactivation of adenylate-uridylate-rich element (ARE)-binding protein BRF1, a target of PI3K-Akt. Consistent decreases in the phosphorylation of Akt and BRF1 were experimentally confirmed following 2 h of hypoxia. These results suggest that the PI3K-Akt signalling pathway might play a role in responses induced by acute hypoxia in skeletal muscles, partially through the dephosphorylation of ARE-binding protein BRF1.


Asunto(s)
Hipoxia/genética , Músculo Esquelético/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Animales , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Hipoxia/metabolismo , Masculino , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/metabolismo , Transducción de Señal
5.
Circ Res ; 116(5): e28-39, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-25605649

RESUMEN

RATIONALE: Sustained activation of Gαq transgenic (Gq) signaling during pressure overload causes cardiac hypertrophy that ultimately progresses to dilated cardiomyopathy. The molecular events that drive hypertrophy decompensation are incompletely understood. Ca(2+)/calmodulin-dependent protein kinase II δ (CaMKIIδ) is activated downstream of Gq, and overexpression of Gq and CaMKIIδ recapitulates hypertrophy decompensation. OBJECTIVE: To determine whether CaMKIIδ contributes to hypertrophy decompensation provoked by Gq. METHODS AND RESULTS: Compared with Gq mice, compound Gq/CaMKIIδ knockout mice developed a similar degree of cardiac hypertrophy but exhibited significantly improved left ventricular function, less cardiac fibrosis and cardiomyocyte apoptosis, and fewer ventricular arrhythmias. Markers of oxidative stress were elevated in mitochondria from Gq versus wild-type mice and respiratory rates were lower; these changes in mitochondrial function were restored by CaMKIIδ deletion. Gq-mediated increases in mitochondrial oxidative stress, compromised membrane potential, and cell death were recapitulated in neonatal rat ventricular myocytes infected with constitutively active Gq and attenuated by CaMKII inhibition. Deep RNA sequencing revealed altered expression of 41 mitochondrial genes in Gq hearts, with normalization of ≈40% of these genes by CaMKIIδ deletion. Uncoupling protein 3 was markedly downregulated in Gq or by Gq expression in neonatal rat ventricular myocytes and reversed by CaMKIIδ deletion or inhibition, as was peroxisome proliferator-activated receptor α. The protective effects of CaMKIIδ inhibition on reactive oxygen species generation and cell death were abrogated by knock down of uncoupling protein 3. Conversely, restoration of uncoupling protein 3 expression attenuated reactive oxygen species generation and cell death induced by CaMKIIδ. Our in vivo studies further demonstrated that pressure overload induced decreases in peroxisome proliferator-activated receptor α and uncoupling protein 3, increases in mitochondrial protein oxidation, and hypertrophy decompensation, which were attenuated by CaMKIIδ deletion. CONCLUSIONS: Mitochondrial gene reprogramming induced by CaMKIIδ emerges as an important mechanism contributing to mitotoxicity in decompensating hypertrophy.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Cardiomegalia/enzimología , Cardiomiopatía Dilatada/etiología , Insuficiencia Cardíaca/etiología , Mitocondrias Cardíacas/fisiología , Acetilcisteína/farmacología , Animales , Apoptosis , Bencilaminas/farmacología , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/deficiencia , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Cardiomegalia/fisiopatología , Cardiomiopatía Dilatada/fisiopatología , Cardiomiopatía Dilatada/prevención & control , Células Cultivadas , Progresión de la Enfermedad , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/deficiencia , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/genética , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/fisiología , Perfilación de la Expresión Génica , Insuficiencia Cardíaca/fisiopatología , Canales Iónicos/biosíntesis , Canales Iónicos/genética , Canales Iónicos/fisiología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Proteínas Mitocondriales/biosíntesis , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/fisiología , Miocitos Cardíacos/metabolismo , Estrés Oxidativo , PPAR alfa/biosíntesis , PPAR alfa/genética , Mutación Puntual , Presión , Interferencia de ARN , ARN Mensajero/biosíntesis , ARN Mensajero/genética , ARN Interferente Pequeño/farmacología , Ratas , Especies Reactivas de Oxígeno , Análisis de Secuencia de ARN , Sulfonamidas/farmacología , Transfección , Proteína Desacopladora 3
6.
Mol Pharmacol ; 88(1): 181-7, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25737495

RESUMEN

G protein-coupled receptors (GPCRs), the largest family of signaling receptors in the human genome, are also the largest class of targets of approved drugs. Are the optimal GPCRs (in terms of efficacy and safety) currently targeted therapeutically? Especially given the large number (∼ 120) of orphan GPCRs (which lack known physiologic agonists), it is likely that previously unrecognized GPCRs, especially orphan receptors, regulate cell function and can be therapeutic targets. Knowledge is limited regarding the diversity and identity of GPCRs that are activated by endogenous ligands and that native cells express. Here, we review approaches to define GPCR expression in tissues and cells and results from studies using these approaches. We identify problems with the available data and suggest future ways to identify and validate the physiologic and therapeutic roles of previously unrecognized GPCRs. We propose that a particularly useful approach to identify functionally important GPCRs with therapeutic potential will be to focus on receptors that show selective increases in expression in diseased cells from patients and experimental animals.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Animales , Regulación de la Expresión Génica , Humanos , Terapia Molecular Dirigida , Receptores Acoplados a Proteínas G/antagonistas & inhibidores , Distribución Tisular
7.
Circ Res ; 112(6): 935-44, 2013 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-23388157

RESUMEN

RATIONALE: Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) has been implicated as a maladaptive mediator of cardiac ischemic injury. We hypothesized that the inflammatory response associated with in vivo ischemia/reperfusion (I/R) is initiated through CaMKII signaling. OBJECTIVE: To assess the contribution of CaMKIIδ to the development of inflammation, infarct, and ventricular dysfunction after in vivo I/R and define early cardiomyocyte-autonomous events regulated by CaMKIIδ using cardiac-specific knockout mice. METHODS AND RESULTS: Wild-type and CaMKIIδ knockout mice were subjected to in vivo I/R by occlusion of the left anterior descending artery for 1 hour followed by reperfusion for various times. CaMKIIδ deletion protected the heart against I/R damage as evidenced by decreased infarct size, attenuated apoptosis, and improved functional recovery. CaMKIIδ deletion also attenuated I/R-induced inflammation and upregulation of nuclear factor-κB (NF-κB) target genes. Further studies demonstrated that I/R rapidly increases CaMKII activity, leading to NF-κB activation within minutes of reperfusion. Experiments using cyclosporine A and cardiac-specific CaMKIIδ knockout mice indicate that NF-κB activation is initiated independent of necrosis and within cardiomyocytes. Expression of activated CaMKII in cardiomyocytes leads to IκB kinase phosphorylation and concomitant increases in nuclear p65. Experiments using an IκB kinase inhibitor support the conclusion that this is a proximal site of CaMKII-mediated NF-κB activation. CONCLUSIONS: This is the first study demonstrating that CaMKIIδ mediates NF-κB activation in cardiomyocytes after in vivo I/R and suggests that CaMKIIδ serves to trigger, as well as to sustain subsequent changes in inflammatory gene expression that contribute to myocardial I/R damage.


Asunto(s)
Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/fisiología , Daño por Reperfusión Miocárdica/etiología , FN-kappa B/metabolismo , Animales , Apoptosis/fisiología , Ciclosporina/farmacología , Perfilación de la Expresión Génica , Corazón , Proteínas I-kappa B/antagonistas & inhibidores , Proteínas I-kappa B/metabolismo , Ratones , Ratones Noqueados , Infarto del Miocardio/patología , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Fosforilación , Recuperación de la Función/fisiología , Factor de Transcripción ReIA/metabolismo , Regulación hacia Arriba
8.
BMC Bioinformatics ; 15: 69, 2014 Mar 12.
Artículo en Inglés | MEDLINE | ID: mdl-24621103

RESUMEN

BACKGROUND: Mandatory deposit of raw microarray data files for public access, prior to study publication, provides significant opportunities to conduct new bioinformatics analyses within and across multiple datasets. Analysis of raw microarray data files (e.g. Affymetrix CEL files) can be time consuming, complex, and requires fundamental computational and bioinformatics skills. The development of analytical workflows to automate these tasks simplifies the processing of, improves the efficiency of, and serves to standardize multiple and sequential analyses. Once installed, workflows facilitate the tedious steps required to run rapid intra- and inter-dataset comparisons. RESULTS: We developed a workflow to facilitate and standardize Meta-Analysis of Affymetrix Microarray Data analysis (MAAMD) in Kepler. Two freely available stand-alone software tools, R and AltAnalyze were embedded in MAAMD. The inputs of MAAMD are user-editable csv files, which contain sample information and parameters describing the locations of input files and required tools. MAAMD was tested by analyzing 4 different GEO datasets from mice and drosophila.MAAMD automates data downloading, data organization, data quality control assesment, differential gene expression analysis, clustering analysis, pathway visualization, gene-set enrichment analysis, and cross-species orthologous-gene comparisons. MAAMD was utilized to identify gene orthologues responding to hypoxia or hyperoxia in both mice and drosophila. The entire set of analyses for 4 datasets (34 total microarrays) finished in ~ one hour. CONCLUSIONS: MAAMD saves time, minimizes the required computer skills, and offers a standardized procedure for users to analyze microarray datasets and make new intra- and inter-dataset comparisons.


Asunto(s)
Biología Computacional/métodos , Bases de Datos Genéticas , Metaanálisis como Asunto , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Programas Informáticos , Animales , Drosophila , Ratones , Control de Calidad
9.
Bioinformatics ; 29(16): 2059-61, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23736530

RESUMEN

SUMMARY: We have developed a web-based query tool, Whole-Genome rVISTA (WGRV), that determines enrichment of transcription factors (TFs) and associated target genes in sets of co-regulated genes. WGRV enables users to query databases containing pre-computed genome coordinates of evolutionarily conserved transcription factor binding sites in the proximal promoters (from 100 bp to 5 kb upstream) of human, mouse and Drosophila genomes. TF binding sites are based on position-weight matrices from the TRANSFAC Professional database. For a given set of co-regulated genes, WGRV returns statistically enriched and evolutionarily conserved binding sites, mapped by the regulatory VISTA (rVISTA) algorithm. Users can then retrieve a list of genes from the query set containing the enriched TF binding sites and their location in the query set promoters. Results are exported in a BED format for rapid visualization in the UCSC genome browser. Flat files of mapped conserved sites and their genomic coordinates are also available for analysis with stand-alone software. AVAILABILITY: http://genome.lbl.gov/cgi-bin/WGRVistaInputCommon.pl.


Asunto(s)
Perfilación de la Expresión Génica , Regiones Promotoras Genéticas , Programas Informáticos , Factores de Transcripción/metabolismo , Algoritmos , Animales , Genómica , Humanos , Internet , Ratones
10.
Am J Physiol Cell Physiol ; 304(10): C927-38, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23392113

RESUMEN

Many common, important diseases are either caused or exacerbated by hyperactivation (e.g., cancer) or inactivation (e.g., heart failure) of the cell division cycle. A better understanding of the cell cycle is critical for interpreting numerous types of physiological changes in cells. Moreover, new insights into how to control it will facilitate new therapeutics for a variety of diseases and new avenues in regenerative medicine. The progression of cells through the four main phases of their division cycle [G(0)/G(1), S (DNA synthesis), G(2), and M (mitosis)] is a highly conserved process orchestrated by several pathways (e.g., transcription, phosphorylation, nuclear import/export, and protein ubiquitination) that coordinate a core cell cycle pathway. This core pathway can also receive inputs that are cell type and cell niche dependent. "Broken cell" methods (e.g., use of labeled nucleotide analogs) to assess for cell cycle activity have revealed important insights regarding the cell cycle but lack the ability to assess living cells in real time (longitudinal studies) and with single-cell resolution. Moreover, such methods often require cell synchronization, which can perturb the pathway under study. Live cell cycle sensors can be used at single-cell resolution in living cells, intact tissue, and whole animals. Use of these more recently available sensors has the potential to reveal physiologically relevant insights regarding the normal and perturbed cell division cycle.


Asunto(s)
Técnicas de Cultivo de Célula , Ciclo Celular/fisiología , División Celular/fisiología , Genes Reporteros , Coloración y Etiquetado , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Replicación del ADN , Humanos , Mitosis , Transducción de Señal
11.
Bioinformatics ; 28(16): 2209-10, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22743224

RESUMEN

UNLABELLED: We introduce GO-Elite, a flexible and powerful pathway analysis tool for a wide array of species, identifiers (IDs), pathways, ontologies and gene sets. In addition to the Gene Ontology (GO), GO-Elite allows the user to perform over-representation analysis on any structured ontology annotations, pathway database or biological IDs (e.g. gene, protein or metabolite). GO-Elite exploits the structured nature of biological ontologies to report a minimal set of non-overlapping terms. The results can be visualized on WikiPathways or as networks. Built-in support is provided for over 60 species and 50 ID systems, covering gene, disease and phenotype ontologies, multiple pathway databases, biomarkers, and transcription factor and microRNA targets. GO-Elite is available as a web interface, GenMAPP-CS plugin and as a cross-platform application. AVAILABILITY: http://www.genmapp.org/go_elite


Asunto(s)
Biología Computacional/métodos , Bases de Datos Genéticas , Almacenamiento y Recuperación de la Información/métodos , Programas Informáticos , Internet , Interfaz Usuario-Computador , Vocabulario Controlado
12.
Proc Natl Acad Sci U S A ; 107(23): 10514-9, 2010 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-20498046

RESUMEN

Two major goals of regenerative medicine are to reproducibly transform adult somatic cells into a pluripotent state and to control their differentiation into specific cell fates. Progress toward these goals would be greatly helped by obtaining a complete picture of the RNA isoforms produced by these cells due to alternative splicing (AS) and alternative promoter selection (APS). To investigate the roles of AS and APS, reciprocal exon-exon junctions were interrogated on a genome-wide scale in differentiating mouse embryonic stem (ES) cells with a prototype Affymetrix microarray. Using a recently released open-source software package named AltAnalyze, we identified 144 genes for 170 putative isoform variants, the majority (67%) of which were predicted to alter protein sequence and domain composition. Verified alternative exons were largely associated with pathways of Wnt signaling and cell-cycle control, and most were conserved between mouse and human. To examine the functional impact of AS, we characterized isoforms for two genes. As predicted by AltAnalyze, we found that alternative isoforms of the gene Serca2 were targeted by distinct microRNAs (miRNA-200b, miRNA-214), suggesting a critical role for AS in cardiac development. Analysis of the Wnt transcription factor Tcf3, using selective knockdown of an ES cell-enriched and characterized isoform, revealed several distinct targets for transcriptional repression (Stmn2, Ccnd2, Atf3, Klf4, Nodal, and Jun) as well as distinct differentiation outcomes in ES cells. The findings herein illustrate a critical role for AS in the specification of ES cells with differentiation, and highlight the utility of global functional analyses of AS.


Asunto(s)
Empalme Alternativo , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Animales , Exones , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Factor 4 Similar a Kruppel , Ratones , MicroARNs/genética , Regiones Promotoras Genéticas , Selección Genética , Transducción de Señal , Transcripción Genética , Proteínas Wnt/metabolismo
13.
J Cell Mol Med ; 16(8): 1656-62, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22225769

RESUMEN

Aging is marked by a decline in LV diastolic function, which encompasses abnormalities in diastolic relaxation, chamber filling and/or passive myocardial stiffness. Genetic tractability and short life span make Drosophila melanogaster an ideal organism to study the effects of aging on heart function, including senescent-associated changes in gene expression and in passive myocardial stiffness. However, use of the Drosophila heart tube to probe deterioration of diastolic performance is subject to at least two challenges: the extent of genetic homology to mammals and the ability to resolve mechanical properties of the bilayered fly heart, which consists of a ventral muscle layer that covers the contractile cardiomyocytes. Here, we argue for widespread use of Drosophila as a novel myocardial aging model by (1) describing diastolic dysfunction in flies, (2) discussing how critical pathways involved in dysfunction are conserved across species and (3) demonstrating the advantage of an atomic force microscopy-based analysis method to measure stiffness of the multilayered Drosophila heart tube versus isolated myocytes from other model systems. By using powerful Drosophila genetic tools, we aim to efficiently alter changes observed in factors that contribute to diastolic dysfunction to understand how one might improve diastolic performance at advanced ages in humans.


Asunto(s)
Diástole/fisiología , Drosophila melanogaster/fisiología , Miocardio/patología , Envejecimiento/patología , Animales , Modelos Animales de Enfermedad
14.
J Biol Chem ; 286(38): 33260-7, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21808067

RESUMEN

The second messenger cAMP is proapoptotic for numerous cell types, but the mechanism for this proapoptotic action is not defined. Here, we use murine CD4(+)/CD8(+) S49 lymphoma cells and isolated thymocytes to assess this mechanism. In WT S49 cells, cAMP acts via protein kinase A (PKA) to induce G(1) phase cell cycle arrest and apoptosis. Treatment of WT and cAMP-Deathless (D-) S49 cells, which lack cAMP-promoted apoptosis, with the PKA agonist 8-(4-chlorophenylthio)-cAMP (CPT-cAMP) differentially regulates transcripts for numerous proapoptotic and antiapoptotic proteins. In contrast, kin-S49 cells (which lack PKA) show no cAMP-promoted changes in transcript expression. In this study, we use knockdown and overexpression approaches to define the role in cAMP/PKA-promoted apoptosis of the proapoptotic factor BIM (Bcl-2 interacting mediator of cell death), whose expression prominently increases in response to CPT-cAMP treatment of WT but not D- or kin- S49 cells. Conditional expression of BimL, one of the three major forms of Bim, increases apoptosis of WT, D-, and kin-S49 cells, whereas inhibition of cAMP-mediated induction of Bim isoforms by shRNAi attenuates CPT-cAMP-mediated apoptosis of WT S49 cells. Bim protein levels increase in subpopulations of CPT-cAMP-treated cells that undergo apoptosis. Thymic CD4(+)/CD8(+) cells isolated from Bim(-/-) mice corroborated the requirement of Bim expression for cAMP-promoted apoptosis. Thus, up-regulation of Bim appears to be an important determinant of cAMP/PKA-mediated apoptosis in immature T cells and may be a mechanism for such apoptosis in other cell types as well.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Apoptosis , Diferenciación Celular , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T/citología , Linfocitos T/enzimología , Animales , Apoptosis/efectos de los fármacos , Proteína 11 Similar a Bcl2 , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/enzimología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/enzimología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , AMP Cíclico/análogos & derivados , AMP Cíclico/farmacología , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Cinética , Ratones , ARN Interferente Pequeño/metabolismo , Linfocitos T/efectos de los fármacos , Tionucleótidos/farmacología , Regulación hacia Arriba/efectos de los fármacos
15.
Exp Physiol ; 97(1): 102-14, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21967898

RESUMEN

The Murphy Roths Large (MRL) mouse, a strain capable of regenerating right ventricular myocardium, has a high postmyocardial infarction (post-MI) survival rate compared with C57BL/6J (C57) mice. The biological processes responsible for this survival advantage are unknown. To assess the effect of genetic background, the LG/J strain, which harbours 75% of the MRL composite genome, was included in the study. The MRL survival advantage versus C57 mice (92 versus 68%, P < 0.05) occurred primarily in the first 5 days; LG/J survival was intermediate (P = n.s.). Microarray data analysis revealed an attenuation of apoptotic (P < 0.05) and stress response transcripts in MRL hearts compared with C57 hearts post-MI. Supporting the microarray results, there were fewer TUNEL-positive cells 1 day post-MI in MRL infarcts compared with C57 infarcts (P = 0.001) and fewer CD45-positive cells in the MRL infarct border zone 2 days post-MI (P < 0.01); the LG/J results were intermediate (P = n.s.). The MRL hearts had smaller infarct scars and attenuated ventricular dilatation 30 days post-MI compared with C57 hearts (P < 0.05). We conclude that the early post-MI survival advantage of MRL mice over the C57 strain is mediated at least in part by reductions in apoptosis and inflammatory infiltration, and that these reductions may influence chronic remodelling. The intermediate survival, apoptosis and inflammation profile of LG/J mice suggests that this high tolerance for MI in the MRL mouse could be derived from its shared genetic background with the LG/J mouse.


Asunto(s)
Apoptosis/genética , Inflamación/genética , Infarto del Miocardio/genética , Remodelación Ventricular/genética , Animales , Dilatación/métodos , Corazón/fisiología , Etiquetado Corte-Fin in Situ/métodos , Antígenos Comunes de Leucocito/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/metabolismo , Tasa de Supervivencia
16.
Am J Physiol Regul Integr Comp Physiol ; 301(3): R727-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21677276

RESUMEN

Stem cells are a potential key strategy for treating neurodegenerative diseases in which the generation of new neurons is critical. A better understanding of the characteristics and molecular properties of neural stem cells (NSCs) and differentiated neurons can help with assessing neuronal maturity and, possibly, in devising better therapeutic strategies. We have performed an in-depth gene expression profiling study of murine NSCs and primary neurons derived from embryonic mouse brains. Microarray analysis revealed a neuron-specific gene expression signature that distinguishes primary neurons from NSCs, with elevated levels of transcripts involved in neuronal functions, such as neurite development and axon guidance in primary neurons and decreased levels of multiple cytokine transcripts. Among the differentially expressed genes, we found a statistically significant enrichment of genes in the ephrin, neurotrophin, CDK5, and actin pathways, which control multiple neuronal-specific functions. We then artificially blocked the cell cycle of NSCs with mitomycin C (MMC) and examined cellular morphology and gene expression signatures. Although these MMC-treated NSCs displayed a neuronal morphology and expressed some neuronal differentiation marker genes, their gene expression patterns were very different from primary neurons. We conclude that 1) fully differentiated mouse primary neurons display a specific neuronal gene expression signature; 2) cell cycle block at the S phase in NSCs with MMC does not induce the formation of fully differentiated neurons; 3) cytokines change their expression pattern during differentiation of NSCs into neurons; and 4) signaling pathways of ephrin, neurotrophin, CDK5, and actin, related to major neuronal features, are dynamically enriched in genes showing changes in expression level.


Asunto(s)
Encéfalo/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Proliferación Celular , Perfilación de la Expresión Génica , Neuronas/metabolismo , Células Madre/metabolismo , Animales , Encéfalo/efectos de los fármacos , Encéfalo/embriología , Ciclo Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , Forma de la Célula/genética , Análisis por Conglomerados , Perfilación de la Expresión Génica/métodos , Regulación del Desarrollo de la Expresión Génica , Redes Reguladoras de Genes , Genotipo , Ratones , Ratones Endogámicos C57BL , Mitomicina/farmacología , Neuronas/efectos de los fármacos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Transducción de Señal/genética , Células Madre/efectos de los fármacos
17.
Cytometry A ; 77(6): 564-70, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20235278

RESUMEN

Genetic based reporters have distinct advantages over classical immunocytochemical techniques for probing cellular functions. Most importantly, they enable dynamic real-time visualization and quantification of cellular processes in living cells and tissue. This study was conducted to generate a genetic based reporter to label cells that transitioned from the G(0) to G(1)/S phases of the cell cycle, hypothesizing that the proximal promoter of the Ki67 (Ki67p) gene, a commonly used cytology marker induced during this transition, would contain the suitable regulatory elements to drive marker gene expression. This study reports the cloning and characterization of the 1.5 kb proximal promoter (Ki67p) of the human Ki67 gene. Ki67p driven GFP expression colocalizes in cells with endogenous Ki67 expression and is correlated with cells transitioning through S/G(2)/M phases of the cell cycle. Treatment Ki67p-GFP expressing HT1080 cells with mitomycin C, an antineoplastic agent, induces P21 and P27 expression, G(1)/S/G(2)M block and attenuates Ki67p activity. Attenuation of the Ki67p also occurs during cell-density induced cell cycle arrest. Taken together, these results indicate that the Ki67p can be used to identify proliferating subpopulations of live cells in intact complex three-dimensional cellular aggregates, such as embryoid bodies, thus providing some unique advantages over conventional immunohistochemical approaches.


Asunto(s)
Biomarcadores/química , Ciclo Celular , Antígeno Ki-67/genética , Regiones Promotoras Genéticas , Animales , Células Cultivadas , Clonación Molecular , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Antígeno Ki-67/química , Ratones , Coloración y Etiquetado
18.
Pharmaceutics ; 12(9)2020 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-32911688

RESUMEN

Non-invasive brain delivery of neurotherapeutics is challenging due to the blood-brain barrier. The revived interest in transferrin receptor antibodies (TfRMAbs) as brain drug-delivery vectors has revealed the effect of dosing regimen, valency, and affinity on brain uptake, TfR expression, and Fc-effector function side effects. These studies have primarily used monovalent TfRMAbs with a human constant region following acute intravenous dosing in mice. The effects of a high-affinity bivalent TfRMAb with a murine constant region, without a fusion partner, following extravascular dosing in mice are, however, not well characterized. Here we elucidate the plasma pharmacokinetics and safety of a high-affinity bivalent TfRMAb with a murine constant region following acute and chronic subcutaneous dosing in adult C57BL/6J male mice. Mice received a single (acute dosing) 3 mg/kg dose, or were treated for four weeks (chronic dosing). TfRMAb and control IgG1 significantly altered reticulocyte counts following acute and chronic dosing, while other hematologic parameters showed minimal change. Chronic TfRMAb dosing did not alter plasma- and brain-iron measurements, nor brain TfR levels, however, it significantly increased splenic-TfR and -iron. Plasma concentrations of TfRMAb were significantly lower in mice chronically treated with IgG1 or TfRMAb. Overall, no injection related reactions were observed in mice.

19.
Pharmacol Res Perspect ; 7(5): e00511, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31463067

RESUMEN

Opioids are powerful analgesics acting via the human µ-opiate receptor (hMOR). Opioid use is associated with adverse effects such as tolerance, addiction, respiratory depression, and constipation. Two synthetic opioids, AH-7921 and U-47700 that were developed in the 1970s but never marketed, have recently appeared on the illegal drug market and in forensic toxicology reports. These agents were initially characterized for their analgesic activity in rodents; however, their pharmacology at hMOR has not been delineated. Thus, we synthesized over 50 chemical analogs based on core AH-7921 and U-47700 structures to assess for their ability to couple to Gαi signaling and induce hMOR internalization. For both the AH-7921 and U-47700 analogs, the 3,4-dichlorobenzoyl substituents were the most potent with comparable EC50 values for inhibition of cAMP accumulation; 26.49 ± 11.2 nmol L-1 and 8.8 ± 4.9 nmol L-1, respectively. Despite similar potencies for Gαi coupling, these two compounds had strikingly different hMOR internalization efficacies: U-47700 (10 µmol L-1) induced ~25% hMOR internalization similar to DAMGO while AH-7921 (10 µmol L-1) induced ~5% hMOR internalization similar to morphine. In addition, the R, R enantiomer of U-47700 is significantly more potent than the S, S enantiomer at hMOR. In conclusion, these data suggest that U-47700 and AH-7921 analogs have high analgesic potential in humans, but with divergent receptor internalization profiles, suggesting that they may exhibit differences in clinical utility or abuse potential.


Asunto(s)
Analgésicos Opioides/síntesis química , Etilenodiaminas/síntesis química , Receptores Opioides mu/metabolismo , Analgésicos Opioides/química , Analgésicos Opioides/farmacología , Línea Celular , AMP Cíclico/metabolismo , Etilenodiaminas/química , Etilenodiaminas/farmacología , Humanos , Estructura Molecular , Receptores Opioides mu/química
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