Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Blood ; 137(14): 1905-1919, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-33751108

RESUMEN

Chromosome 13q deletion [del(13q)], harboring the miR-15a/16-1 cluster, is one of the most common genetic alterations in mature B-cell malignancies, which originate from germinal center (GC) and post-GC B cells. Moreover, miR-15a/16 expression is frequently reduced in lymphoma and multiple myeloma (MM) cells without del(13q), suggesting important tumor-suppressor activity. However, the role of miR-15a/16-1 in B-cell activation and initiation of mature B-cell neoplasms remains to be determined. We show that conditional deletion of the miR-15a/16-1 cluster in murine GC B cells induces moderate but widespread molecular and functional changes including an increased number of GC B cells, percentage of dark zone B cells, and maturation into plasma cells. With time, this leads to development of mature B-cell neoplasms resembling human extramedullary plasmacytoma (EP) as well as follicular and diffuse large B-cell lymphomas. The indolent nature and lack of bone marrow involvement of EP in our murine model resembles human primary EP rather than MM that has progressed to extramedullary disease. We corroborate human primary EP having low levels of miR-15a/16 expression, with del(13q) being the most common genetic loss. Additionally, we show that, although the mutational profile of human EP is similar to MM, there are some exceptions such as the low frequency of hyperdiploidy in EP, which could account for different disease presentation. Taken together, our studies highlight the significant role of the miR-15a/16-1 cluster in the regulation of the GC reaction and its fundamental context-dependent tumor-suppression function in plasma cell and B-cell malignancies.


Asunto(s)
Linfoma de Células B Grandes Difuso/genética , MicroARNs/genética , Neoplasias de Células Plasmáticas/genética , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Deleción Cromosómica , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/patología , Cromosomas Humanos Par 13/genética , Eliminación de Gen , Regulación Neoplásica de la Expresión Génica , Humanos , Linfoma de Células B Grandes Difuso/patología , Ratones Endogámicos C57BL , Familia de Multigenes , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Neoplasias de Células Plasmáticas/patología , Células Plasmáticas/metabolismo , Células Plasmáticas/patología , Plasmacitoma/genética , Plasmacitoma/patología
2.
Proteomics ; 22(9): e2100265, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35094488

RESUMEN

Pluripotent stem cells (PSC) endocrine differentiation at a large scale allows sampling of transcriptome and proteome with phosphoproteome (proteoform) at specific time points. We describe the dynamic time course of changes in cells undergoing directed beta-cell differentiation and show target proteins or previously unknown phosphorylation of critical proteins in pancreas development, NKX6-1, and Chromogranin A (CHGA). We describe fluctuations in the correlation between gene expression, protein abundance, and phosphorylation, following differentiation protocol perturbations at all stages to identify proteoform profiles. Our modeling recognizes outliers on a phenomic landscape of endocrine differentiation, and we describe new biological pathways involved. We have validated our proteomic data by analyzing independent single-cell RNAseq datasets for in-vitro pancreatic islet production and corroborated our findings for several proteins suggestive as targets for future research. The single-cell analysis combined with proteoform data places new protein targets within the specific time point and at the specific pancreatic lineage of differentiating stem cells. We suggest that non-correlating proteins abundances or new phosphorylation motifs of NKX6.1 and CHGA point to new signaling pathways that may play an essential role in beta-cell development. We present our findings for the research community's use to improve endocrine differentiation protocols and developmental studies.


Asunto(s)
Células Madre Embrionarias Humanas , Células Madre Pluripotentes , Diferenciación Celular/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Humanos , Células Madre Pluripotentes/metabolismo , Proteómica
3.
J Immunol ; 195(9): 4185-97, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26416268

RESUMEN

Virus-specific CD8(+) T cells expand dramatically during acute EBV infection, and their persistence is important for lifelong control of EBV-related disease. To better define the generation and maintenance of these effective CD8(+) T cell responses, we used microarrays to characterize gene expression in total and EBV-specific CD8(+) T cells isolated from the peripheral blood of 10 individuals followed from acute infectious mononucleosis (AIM) into convalescence (CONV). In total CD8(+) T cells, differential expression of genes in AIM and CONV was most pronounced among those encoding proteins important in T cell activation/differentiation, cell division/metabolism, chemokines/cytokines and receptors, signaling and transcription factors (TF), immune effector functions, and negative regulators. Within these categories, we identified 28 genes that correlated with CD8(+) T cell expansion in response to an acute EBV infection. In EBV-specific CD8(+) T cells, we identified 33 genes that were differentially expressed in AIM and CONV. Two important TF, T-bet and eomesodermin, were upregulated and maintained at similar levels in both AIM and CONV; in contrast, protein expression declined from AIM to CONV. Expression of these TF varied among cells with different epitope specificities. Collectively, gene and protein expression patterns suggest that a large proportion, if not a majority of CD8(+) T cells in AIM are virus specific, activated, dividing, and primed to exert effector activities. High expression of T-bet and eomesodermin may help to maintain effector mechanisms in activated cells and to enable proliferation and transition to earlier differentiation states in CONV.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Herpesvirus Humano 4/inmunología , Mononucleosis Infecciosa/inmunología , Transcriptoma , ADP-Ribosil Ciclasa 1/genética , Enfermedad Aguda , Adolescente , Adulto , Femenino , Humanos , Masculino , Receptores de Interleucina-7/genética , Factores de Transcripción/genética
4.
J Biol Chem ; 289(26): 18327-38, 2014 Jun 27.
Artículo en Inglés | MEDLINE | ID: mdl-24778188

RESUMEN

Adipose tissue expansion requires growth and proliferation of adipocytes and the concomitant expansion of their stromovascular network. We have used an ex vivo angiogenesis assay to study the mechanisms involved in adipose tissue expansion. In this assay, adipose tissue fragments placed under pro-angiogenic conditions form sprouts composed of endothelial, perivascular, and other proliferative cells. We find that sprouting was directly stimulated by insulin and was enhanced by prior treatment of mice with the insulin sensitizer rosiglitazone. Moreover, basal and insulin-stimulated sprouting increased progressively over 30 weeks of high fat diet feeding, correlating with tissue expansion during this period. cDNA microarrays analyzed to identify genes correlating with insulin-stimulated sprouting surprisingly revealed only four positively correlating (Fads3, Tmsb10, Depdc6, and Rasl12) and four negatively correlating (Asph, IGFbp4, Ppm1b, and Adcyap1r1) genes. Among the proteins encoded by these genes, IGFbp4, which suppresses IGF-1 signaling, has been previously implicated in angiogenesis, suggesting a role for IGF-1 in adipose tissue expandability. Indeed, IGF-1 potently stimulated sprouting, and the presence of activated IGF-1 receptors in the vasculature was revealed by immunostaining. Recombinant IGFbp4 blocked the effects of insulin and IGF-1 on mouse adipose tissue sprouting and also suppressed sprouting from human subcutaneous adipose tissue. These results reveal an important role of IGF-1/IGFbp4 signaling in post-developmental adipose tissue expansion.


Asunto(s)
Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Proliferación Celular , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Obesidad/metabolismo , Animales , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/metabolismo , Humanos , Técnicas In Vitro , Proteína 4 de Unión a Factor de Crecimiento Similar a la Insulina/genética , Factor I del Crecimiento Similar a la Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/genética , Obesidad/fisiopatología , Transducción de Señal
5.
Mol Omics ; 20(1): 6-18, 2024 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-37681418

RESUMEN

Single-cell analysis has clearly established itself in biology and biomedical fields as an invaluable tool that allows one to comprehensively understand the relationship between cells, including their types, states, transitions, trajectories, and spatial position. Scientific methods such as fluorescence labeling, nanoscale super-resolution microscopy, advances in single cell RNAseq and proteomics technologies, provide more detailed information about biological processes which were not evident with the analysis of bulk material. This new era of single-cell biology provides a better understanding of such complex biological systems as cancer, inflammation, immunity mechanism and aging processes, and opens the door into the field of drug response heterogeneity. The latest discoveries of cellular heterogeneity gives us a unique understanding of complex biological processes, such as disease mechanism, and will lead to new strategies for better and personalized treatment strategies. Recently, single-cell proteomics techniques that allow quantification of thousands of proteins from single mammalian cells have been introduced. Here we present an improved single-cell mass spectrometry-based proteomics platform called SCREEN (Single Cell pRotEomE aNalysis) for deep and high-throughput single-cell proteome coverage with high efficiency, less turnaround time and with an improved ability for protein quantitation across more cells than previously achieved. We applied this new platform to analyze the single-cell proteomic landscape under different drug treatment over time to uncover heterogeneity in cancer cell response, which for the first time, to our knowledge, has been achieved by mass spectrometry based analytical methods. We discuss challenges in single-cell proteomics, future improvements and general trends with the goal to encourage forthcoming technical developments.


Asunto(s)
Proteoma , Proteómica , Animales , Proteoma/metabolismo , Proteómica/métodos , Espectrometría de Masas/métodos , Análisis de la Célula Individual , Mamíferos/metabolismo
6.
J Lipid Res ; 54(10): 2697-707, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23924694

RESUMEN

Adipose tissue lipogenesis is paradoxically impaired in human obesity, promoting ectopic triglyceride (TG) deposition, lipotoxicity, and insulin resistance. We previously identified mitogen-activated protein kinase kinase kinase kinase 4 (Map4k4), a sterile 20 protein kinase reported to be upstream of c-Jun NH2-terminal kinase (JNK) signaling, as a novel negative regulator of insulin-stimulated glucose transport in adipocytes. Using full-genome microarray analysis we uncovered a novel role for Map4k4 as a suppressor of lipid synthesis. We further report here the surprising finding that Map4k4 suppresses adipocyte lipogenesis independently of JNK. Thus, while Map4k4 silencing in adipocytes enhances the expression of lipogenic enzymes, concomitant with increased conversion of (14)C-glucose and (14)C-acetate into TGs and fatty acids, JNK1 and JNK2 depletion causes the opposite effects. Furthermore, high expression of Map4k4 fails to activate endogenous JNK, while Map4k4 depletion does not attenuate JNK activation by tumor necrosis factor α. Map4k4 silencing in cultured adipocytes elevates both the total protein expression and cleavage of sterol-regulated element binding protein-1 (Srebp-1) in a rapamycin-sensitive manner, consistent with Map4k4 signaling via mechanistic target of rapamycin complex 1 (mTORC1). We show Map4k4 depletion requires Srebp-1 upregulation to increase lipogenesis and further show that Map4k4 promotes AMP-protein kinase (AMPK) signaling and the phosphorylation of mTORC1 binding partner raptor (Ser792) to inhibit mTORC1. Our results indicate that Map4k4 inhibits adipose lipogenesis by suppression of Srebp-1 in an AMPK- and mTOR-dependent but JNK-independent mechanism.


Asunto(s)
Adipocitos/metabolismo , Lipogénesis , Sistema de Señalización de MAP Quinasas , Proteínas Serina-Treonina Quinasas/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Activación Enzimática , Expresión Génica , Técnicas de Silenciamiento del Gen , Ratones , Obesidad/enzimología , Serina-Treonina Quinasas TOR/metabolismo , Activación Transcripcional , Triglicéridos/biosíntesis , Quinasa de Factor Nuclear kappa B
7.
bioRxiv ; 2023 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-37609178

RESUMEN

How ubiquitous circadian clocks orchestrate tissue-specific outputs is not well understood. Pancreatic ß cell-autonomous clocks attune insulin secretion to daily energy cycles, and desynchrony from genetic or behavioral disruptions raises type 2 diabetes risk. We show that the transcription factor DEC1, a clock component induced in adult ß cells, coordinates their glucose responsiveness by synchronizing energy metabolism and secretory gene oscillations. Dec1-ablated mice develop lifelong hypo-insulinemic diabetes, despite normal islet formation and intact circadian Clock and Bmal1 activators. DEC1, but not CLOCK/BMAL1, binds maturity-linked genes that mediate respiratory metabolism and insulin exocytosis, and Dec1 loss disrupts their transcription synchrony. Accordingly, ß-cell Dec1 ablation causes hypo-insulinemia due to immature glucose responsiveness, dampening insulin rhythms. Thus, Dec1 links circadian clockwork to the ß-cell maturation process, aligning metabolism to diurnal energy cycles.

8.
Dev Biol ; 358(2): 356-67, 2011 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-21787765

RESUMEN

Segmentation of the vertebrate hindbrain into multiple rhombomeres is essential for proper formation of the cerebellum, cranial nerves and cranial neural crest. Paralog group 1 (PG1) hox genes are expressed early in the caudal hindbrain and are required for rhombomere formation. Accordingly, loss of PG1 hox function disrupts development of caudal rhombomeres in model organisms and causes brainstem defects, associated with cognitive impairment, in humans. In spite of this important role for PG1 hox genes, transcriptional targets of PG1 proteins are not well characterized. Here we use ectopic expression together with embryonic dissection to identify novel targets of the zebrafish PG1 gene hoxb1b. Of 100 genes up-regulated by hoxb1b, 54 were examined and 25 were found to represent novel hoxb1b regulated hindbrain genes. The ppp1r14al gene was analyzed in greater detail and our results indicate that Hoxb1b is likely to directly regulate ppp1r14al expression in rhombomere 4. Furthermore, ppp1r14al is essential for establishment of the earliest hindbrain signaling-center in rhombomere 4 by regulating expression of fgf3.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Proteínas de Homeodominio/genética , Fosfoproteínas Fosfatasas/genética , Rombencéfalo/embriología , Rombencéfalo/metabolismo , Proteínas de Pez Cebra/genética , Animales , Secuencia de Bases , Cartilla de ADN/genética , Factor 3 de Crecimiento de Fibroblastos/antagonistas & inhibidores , Factor 3 de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Regulación del Desarrollo de la Expresión Génica , Factor Nuclear 1-beta del Hepatocito/genética , Proteínas de Homeodominio/antagonistas & inhibidores , Hibridación in Situ , Factor de Transcripción MafB/genética , Proteínas del Tejido Nervioso/genética , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/metabolismo , Filogenia , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/genética , Transducción de Señal , Transcriptoma , Pez Cebra/embriología , Pez Cebra/genética , Proteínas de Pez Cebra/antagonistas & inhibidores , Proteínas de Pez Cebra/metabolismo
9.
Circulation ; 123(2): 186-94, 2011 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-21200001

RESUMEN

BACKGROUND: Adipose tissue expands in response to excess caloric intake, but individuals prone to deposit visceral instead of subcutaneous adipose tissue have higher risk of metabolic disease. The role of angiogenesis in the expandability of human adipose tissue depots is unknown. The objective of this study was to measure angiogenesis in visceral and subcutaneous adipose tissue and to establish whether there is a relationship between obesity, metabolic status, and the angiogenic properties of these depots. METHODS AND RESULTS: Angiogenic capacity was determined by quantifying capillary branch formation from human adipose tissue explants embedded in Matrigel, and capillary density was assessed by immunohistochemistry. Subcutaneous adipose tissue had a greater angiogenic capacity than visceral tissue, even after normalization to its higher initial capillary density. Gene array analyses revealed significant differences in expression of angiogenic genes between depots, including an increased subcutaneous expression of angiopoietin-like protein 4, which is proangiogenic in an adipose tissue context. Subcutaneous capillary density and angiogenic capacity decreased with morbid obesity, and subcutaneous, but not visceral, adipose tissue angiogenic capacity correlated negatively with insulin sensitivity. CONCLUSIONS: These data imply that subcutaneous adipose tissue has a higher capacity to expand its capillary network than visceral tissue, but this capacity decreases with morbid obesity. The decrease correlates with insulin resistance, suggesting that impairment of subcutaneous adipose tissue angiogenesis may contribute to metabolic disease pathogenesis.


Asunto(s)
Grasa Intraabdominal/irrigación sanguínea , Neovascularización Patológica/fisiopatología , Neovascularización Fisiológica/fisiología , Obesidad/fisiopatología , Grasa Subcutánea/irrigación sanguínea , Adulto , Proteína 4 Similar a la Angiopoyetina , Angiopoyetinas/metabolismo , Índice de Masa Corporal , Derivación Gástrica , Humanos , Resistencia a la Insulina/fisiología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/fisiopatología , Persona de Mediana Edad , Obesidad/metabolismo , Obesidad/cirugía , Grasa Subcutánea/metabolismo , Grasa Subcutánea/fisiopatología
10.
Artículo en Inglés | MEDLINE | ID: mdl-36376065

RESUMEN

A family, with two affected identical twins with early-onset recessive inherited retinal degeneration, was analyzed to determine the underlying genetic cause of pathology. Exome sequencing revealed a rare and previously reported causative variant (c.1923_1969delinsTCTGGG; p.Asn643Glyfs*29) in the PDE6B gene in the affected twins and their unaffected father. Further investigation, using genome sequencing, identified a novel ∼7.5-kb deletion (Chr 4:670,405-677,862del) encompassing the ATP5ME gene, part of the 5' UTR of MYL5, and a 378-bp (Chr 4:670,405-670,782) region from the 3' UTR of PDE6B in the affected twins and their unaffected mother. Both variants segregated with disease in the family. Analysis of the relative expression of PDE6B, in peripheral blood cells, also revealed a significantly lower level of PDE6B transcript in affected siblings compared to a normal control. PDE6B is associated with recessive rod-cone degeneration and autosomal dominant congenital stationary night blindness. Ophthalmic evaluation of these patients showed night blindness, fundus abnormalities, and peripheral vision loss, which are consistent with PDE6B-associated recessive retinal degeneration. These findings suggest that the loss of PDE6B transcript resulting from the compound heterozygous pathogenic variants is the underlying cause of recessive rod-cone degeneration in the study family.


Asunto(s)
Ceguera Nocturna , Degeneración Retiniana , Humanos , Degeneración Retiniana/genética , Mutación del Sistema de Lectura/genética , Ceguera Nocturna/genética , Ceguera/genética , Mutación INDEL , Linaje , Mutación , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 6/genética
11.
J Neurosci ; 30(21): 7152-67, 2010 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-20505083

RESUMEN

Histone methyltransferases specific for the histone H3-lysine 9 residue, including Setdb1 (Set domain, bifurcated 1)/Eset/Kmt1e are associated with repressive chromatin remodeling and expressed in adult brain, but potential effects on neuronal function and behavior remain unexplored. Here, we report that transgenic mice with increased Setdb1 expression in adult forebrain neurons show antidepressant-like phenotypes in behavioral paradigms for anhedonia, despair, and learned helplessness. Chromatin immunoprecipitation in conjunction with DNA tiling arrays (ChIP-chip) revealed that genomic occupancies of neuronal Setdb1 are limited to <1% of annotated genes, which include the NMDA receptor subunit NR2B/Grin2B and other ionotropic glutamate receptor genes. Chromatin conformation capture and Setdb1-ChIP revealed a loop formation tethering the NR2B/Grin2b promoter to the Setdb1 target site positioned 30 kb downstream of the transcription start site. In hippocampus and ventral striatum, two key structures in the neuronal circuitry regulating mood-related behaviors, Setdb1-mediated repressive histone methylation at NR2B/Grin2b was associated with decreased NR2B expression and EPSP insensitivity to pharmacological blockade of NR2B, and accelerated NMDA receptor desensitization consistent with a shift in NR2A/B subunit ratios. In wild-type mice, systemic treatment with the NR2B antagonist, Ro25-6981 [R-(R,S)-alpha-(4-hydroxyphenyl)-beta-methyl-4-(phenylmethyl)-1-piperidine propranol], and hippocampal small interfering RNA-mediated NR2B/Grin2b knockdown resulted in behavioral changes similar to those elicited by the Setdb1 transgene. Together, these findings point to a role for neuronal Setdb1 in the regulation of affective and motivational behaviors through repressive chromatin remodeling at a select set of target genes, resulting in altered NMDA receptor subunit composition and other molecular adaptations.


Asunto(s)
Afecto/fisiología , Conducta Animal/fisiología , Regulación de la Expresión Génica/fisiología , Proteína Metiltransferasas/metabolismo , Receptores de N-Metil-D-Aspartato/metabolismo , Adaptación Ocular/efectos de los fármacos , Adaptación Ocular/genética , Afecto/efectos de los fármacos , Factores de Edad , Animales , Animales Recién Nacidos , Reacción de Prevención/efectos de los fármacos , Reacción de Prevención/fisiología , Conducta Animal/efectos de los fármacos , Proteína Quinasa Tipo 2 Dependiente de Calcio Calmodulina/genética , Células Cultivadas , Cromatina/metabolismo , Inmunoprecipitación de Cromatina/métodos , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Electrochoque/efectos adversos , Fármacos actuantes sobre Aminoácidos Excitadores/farmacología , Potenciales Postsinápticos Excitadores/efectos de los fármacos , Potenciales Postsinápticos Excitadores/genética , Conducta Exploratoria/efectos de los fármacos , Conducta Exploratoria/fisiología , Miedo/efectos de los fármacos , Miedo/fisiología , Preferencias Alimentarias/efectos de los fármacos , Preferencias Alimentarias/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Proteínas Fluorescentes Verdes/genética , Hipocampo/citología , N-Metiltransferasa de Histona-Lisina , Humanos , Pérdida de Tono Postural/efectos de los fármacos , Pérdida de Tono Postural/fisiología , Aprendizaje por Laberinto/efectos de los fármacos , Aprendizaje por Laberinto/fisiología , Potenciales de la Membrana/efectos de los fármacos , Potenciales de la Membrana/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Actividad Motora/efectos de los fármacos , Actividad Motora/genética , Neuronas/efectos de los fármacos , Neuronas/fisiología , Neuronas/ultraestructura , Técnicas de Placa-Clamp/métodos , Proteína Metiltransferasas/genética , ARN Interferente Pequeño/farmacología , Receptores de N-Metil-D-Aspartato/genética , Sacarosa/administración & dosificación
12.
J Biol Chem ; 285(36): 27581-9, 2010 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-20587420

RESUMEN

Protein kinase B/Akt protein kinases control an array of diverse functions, including cell growth, survival, proliferation, and metabolism. We report here the identification of pleckstrin homology-like domain family B member 1 (PHLDB1) as an insulin-responsive protein that enhances Akt activation. PHLDB1 contains a pleckstrin homology domain, which we show binds phosphatidylinositol PI(3,4)P(2), PI(3,5)P(2), and PI(3,4,5)P(3), as well as a Forkhead-associated domain and coiled coil regions. PHLDB1 expression is increased during adipocyte differentiation, and it is abundant in many mouse tissues. Both endogenous and HA- or GFP-tagged PHLDB1 displayed a cytoplasmic disposition in unstimulated cultured adipocytes but translocated to the plasma membrane in response to insulin. Depletion of PHLDB1 by siRNA inhibited insulin stimulation of Akt phosphorylation but not tyrosine phosphorylation of IRS-1. RNAi-based silencing of PHLDB1 in cultured adipocytes also attenuated insulin-stimulated deoxyglucose transport and Myc-GLUT4-EGFP translocation to the plasma membrane, whereas knockdown of the PHLDB1 isoform PHLDB2 failed to attenuate insulin-stimulated deoxyglucose transport. Furthermore, adenovirus-mediated expression of PHLDB1 in adipocytes enhanced insulin-stimulated Akt and p70 S6 kinase phosphorylation, as well as GLUT4 translocation. These results indicate that PHLDB1 is a novel modulator of Akt protein kinase activation by insulin.


Asunto(s)
Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Transportador de Glucosa de Tipo 4/metabolismo , Insulina/farmacología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células 3T3-L1 , Animales , Proteínas Sanguíneas/química , Activación Enzimática/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Silenciador del Gen , Glucosa/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Ratones , Fosfatos de Fosfatidilinositol/metabolismo , Fosfoproteínas/química , Fosforilación/efectos de los fármacos , Estructura Terciaria de Proteína , Transporte de Proteínas/efectos de los fármacos , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Homología de Secuencia de Aminoácido
13.
Am J Physiol Heart Circ Physiol ; 301(4): H1425-37, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21765057

RESUMEN

Thoracic perivascular adipose tissue (PVAT) is a unique adipose depot that likely influences vascular function and susceptibility to pathogenesis in obesity and the metabolic syndrome. Surprisingly, PVAT has been reported to share characteristics of both brown and white adipose, but a detailed direct comparison to interscapular brown adipose tissue (BAT) has not been performed. Here we show by full genome DNA microarray analysis that global gene expression profiles of PVAT are virtually identical to BAT, with equally high expression of Ucp-1, Cidea, and other genes known to be uniquely or very highly expressed in BAT. PVAT and BAT also displayed nearly identical phenotypes upon immunohistochemical analysis, and electron microscopy confirmed that PVAT contained multilocular lipid droplets and abundant mitochondria. Compared with white adipose tissue (WAT), PVAT and BAT from C57BL6/J mice fed a high-fat diet for 13 wk had markedly lower expression of immune cell-enriched mRNAs, suggesting resistance to obesity-induced inflammation. Indeed, staining of BAT and PVAT for macrophage markers (F4/80 and CD68) in obese mice showed virtually no macrophage infiltration, and FACS analysis of BAT confirmed the presence of very few CD11b(+)/CD11c(+) macrophages in BAT (1.0%) compared with WAT (31%). In summary, murine PVAT from the thoracic aorta is virtually identical to interscapular BAT, is resistant to diet-induced macrophage infiltration, and thus may play an important role in protecting the vascular bed from inflammatory stress.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Tejido Adiposo/fisiología , Vasos Sanguíneos/fisiología , Dieta/efectos adversos , Inflamación/inducido químicamente , Inflamación/prevención & control , Tejido Adiposo/ultraestructura , Tejido Adiposo Pardo/ultraestructura , Animales , Aorta Torácica/metabolismo , Proteínas Reguladoras de la Apoptosis/biosíntesis , Vasos Sanguíneos/ultraestructura , Grasas de la Dieta/efectos adversos , Citometría de Flujo , Inmunohistoquímica , Insulina/sangre , Canales Iónicos/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis por Micromatrices , Microscopía Electrónica de Transmisión , Mitocondrias Cardíacas/fisiología , Proteínas Mitocondriales/biosíntesis , Infiltración Neutrófila/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteína Desacopladora 1
14.
Proc Natl Acad Sci U S A ; 105(22): 7833-8, 2008 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-18509062

RESUMEN

Storage of energy as triglyceride in large adipose-specific lipid droplets is a fundamental need in all mammals. Efficient sequestration of fat in adipocytes also prevents fatty acid overload in skeletal muscle and liver, which can impair insulin signaling. Here we report that the Cide domain-containing protein Cidea, previously thought to be a mitochondrial protein, colocalizes around lipid droplets with perilipin, a regulator of lipolysis. Cidea-GFP greatly enhances lipid droplet size when ectopically expressed in preadipocytes or COS cells. These results explain previous findings showing that depletion of Cidea with RNAi markedly elevates lipolysis in human adipocytes. Like perilipin, Cidea and the related lipid droplet protein Cidec/FSP27 are controlled by peroxisome proliferator-activated receptor gamma (PPARgamma). Treatment of lean or obese mice with the PPARgamma agonist rosiglitazone markedly up-regulates Cidea expression in white adipose tissue (WAT), increasing lipid deposition. Strikingly, in both omental and s.c. WAT from BMI-matched obese humans, expression of Cidea, Cidec/FSP27, and perilipin correlates positively with insulin sensitivity (HOMA-IR index). Thus, Cidea and other lipid droplet proteins define a novel, highly regulated pathway of triglyceride deposition in human WAT. The data support a model whereby failure of this pathway results in ectopic lipid accumulation, insulin resistance, and its associated comorbidities in humans.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Resistencia a la Insulina , Triglicéridos/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo Blanco/citología , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/análisis , Proteínas Reguladoras de la Apoptosis/genética , Índice de Masa Corporal , Proteínas Portadoras , Humanos , Lipólisis , Masculino , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Obesidad/metabolismo , PPAR gamma/agonistas , PPAR gamma/genética , PPAR gamma/metabolismo , Perilipina-1 , Fosfoproteínas/análisis , Fosfoproteínas/metabolismo , Proteínas/genética , Proteínas/metabolismo , Interferencia de ARN , ARN Mensajero/metabolismo , Rosiglitazona , Tiazolidinedionas/farmacología
15.
Cell Stem Cell ; 26(1): 108-122.e10, 2020 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-31839570

RESUMEN

Stem-cell-derived tissues could transform disease research and therapy, yet most methods generate functionally immature products. We investigate how human pluripotent stem cells (hPSCs) differentiate into pancreatic islets in vitro by profiling DNA methylation, chromatin accessibility, and histone modification changes. We find that enhancer potential is reset upon lineage commitment and show how pervasive epigenetic priming steers endocrine cell fates. Modeling islet differentiation and maturation regulatory circuits reveals genes critical for generating endocrine cells and identifies circadian control as limiting for in vitro islet function. Entrainment to circadian feeding/fasting cycles triggers islet metabolic maturation by inducing cyclic synthesis of energy metabolism and insulin secretion effectors, including antiphasic insulin and glucagon pulses. Following entrainment, hPSC-derived islets gain persistent chromatin changes and rhythmic insulin responses with a raised glucose threshold, a hallmark of functional maturity, and function within days of transplantation. Thus, hPSC-derived tissues are amenable to functional improvement by circadian modulation.


Asunto(s)
Diferenciación Celular , Ritmo Circadiano , Islotes Pancreáticos/citología , Células Madre Pluripotentes/citología , Glucagón/metabolismo , Humanos , Insulina/metabolismo , Secreción de Insulina , Islotes Pancreáticos/metabolismo
16.
Cell Metab ; 31(5): 1004-1016.e5, 2020 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-32375022

RESUMEN

A drastic transition at birth, from constant maternal nutrient supply in utero to intermittent postnatal feeding, requires changes in the metabolic system of the neonate. Despite their central role in metabolic homeostasis, little is known about how pancreatic ß cells adjust to the new nutritional challenge. Here, we find that after birth ß cell function shifts from amino acid- to glucose-stimulated insulin secretion in correlation with the change in the nutritional environment. This adaptation is mediated by a transition in nutrient sensitivity of the mTORC1 pathway, which leads to intermittent mTORC1 activity. Disrupting nutrient sensitivity of mTORC1 in mature ß cells reverts insulin secretion to a functionally immature state. Finally, manipulating nutrient sensitivity of mTORC1 in stem cell-derived ß cells in vitro strongly enhances their glucose-responsive insulin secretion. These results reveal a mechanism by which nutrients regulate ß cell function, thereby enabling a metabolic adaptation for the newborn.


Asunto(s)
Glucosa/metabolismo , Nutrientes/metabolismo , Animales , Células Cultivadas , Humanos , Secreción de Insulina , Ratones , Ratones Endogámicos C57BL
17.
Sci Transl Med ; 12(565)2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33055246

RESUMEN

Extracellular vesicles (EVs) derived from various stem cell sources induce cardioprotective effects during ischemia-reperfusion injury (IRI). These have been attributed mainly to the antiapoptotic, proangiogenic, microRNA (miRNA) cargo within the stem cell-derived EVs. However, the mechanisms of EV-mediated endothelial signaling to cardiomyocytes, as well as their therapeutic potential toward ischemic myocardial injury, are not clear. EV content beyond miRNA that may contribute to cardioprotection has not been fully illuminated. This study characterized the protein cargo of human vascular endothelial EVs (EEVs) to identify lead cardioactive proteins and assessed the effect of EEVs on human laminar cardiac tissues (hlCTs) exposed to IRI. We mapped the protein content of human vascular EEVs and identified proteins that were previously associated with cellular metabolism, redox state, and calcium handling, among other processes. Analysis of the protein landscape of human cardiomyocytes revealed corresponding modifications induced by EEV treatment. To assess their human-specific cardioprotection in vitro, we developed a human heart-on-a-chip IRI assay using human stem cell-derived, engineered cardiac tissues. We found that EEVs alleviated cardiac cell death as well as the loss in contractile capacity during and after simulated IRI in an uptake- and dose-dependent manner. Moreover, we found that EEVs increased the respiratory capacity of normoxic cardiomyocytes. These results suggest that vascular EEVs rescue hlCTs exposed to IRI possibly by supplementing injured myocytes with cargo that supports multiple metabolic and salvage pathways and therefore may serve as a multitargeted therapy for IRI.


Asunto(s)
Vesículas Extracelulares , MicroARNs , Daño por Reperfusión , Apoptosis , Humanos , Miocitos Cardíacos
18.
J Clin Invest ; 116(1): 125-36, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16374519

RESUMEN

Using an siRNA-based screen, we identified the transcriptional corepressor RIP140 as a negative regulator of insulin-responsive hexose uptake and oxidative metabolism in 3T3-L1 adipocytes. Affymetrix GeneChip profiling revealed that RIP140 depletion upregulates the expression of clusters of genes in the pathways of glucose uptake, glycolysis, TCA cycle, fatty acid oxidation, mitochondrial biogenesis, and oxidative phosphorylation in these cells. Conversely, we show that reexpression of RIP140 in mouse embryonic fibroblasts derived from RIP140-null mice downregulates expression of many of these same genes. Consistent with these microarray data, RIP140 gene silencing in cultured adipocytes increased both conversion of [14C]glucose to CO2 and mitochondrial oxygen consumption. RIP140-null mice, previously reported to resist weight gain on a high-fat diet, are shown here to display enhanced glucose tolerance and enhanced responsiveness to insulin compared with matched wild-type mice upon high-fat feeding. Mechanistically, RIP140 was found to require the nuclear receptor ERRalpha to regulate hexose uptake and mitochondrial proteins SDHB and CoxVb, although it likely acts through other nuclear receptors as well. We conclude that RIP140 is a major suppressor of adipocyte oxidative metabolism and mitochondrial biogenesis, as well as a negative regulator of whole-body glucose tolerance and energy expenditure in mice.


Asunto(s)
Adipocitos/metabolismo , Mitocondrias/fisiología , Proteínas Nucleares/metabolismo , Fosforilación Oxidativa , Células 3T3 , Proteínas Adaptadoras Transductoras de Señales , Animales , Ciclo del Ácido Cítrico/fisiología , Metabolismo Energético , Glucosa/metabolismo , Glucólisis/fisiología , Ratones , Proteína de Interacción con Receptores Nucleares 1 , Proteínas Represoras/metabolismo
19.
Cell Rep ; 27(8): 2281-2291.e5, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-31116975

RESUMEN

In vitro differentiation of pluripotent cells into ß cells is a promising alternative to cadaveric-islet transplantation as a cure for type 1 diabetes (T1D). During the directed differentiation of human embryonic stem cells (hESCS) by exogenous factors, numerous genes that affect the differentiation process are turned on and off autonomously. Manipulating these reactions could increase the efficiency of differentiation and provide a more complete control over the final composition of cell populations. To uncover in vitro autonomous responses, we performed single-cell RNA sequencing on hESCs as they differentiate in spherical clusters. We observed that endocrine cells and their progenitors exist beside one another in separate compartments that activate distinct genetic pathways. WNT pathway inhibition in the endocrine domain of the differentiating clusters reveals a necessary role for the WNT inhibitor APC during islet formation in vivo. Accordingly, WNT inhibition in vitro causes an increase in the proportion of differentiated endocrine cells.


Asunto(s)
Páncreas/crecimiento & desarrollo , Páncreas/metabolismo , Células Madre/metabolismo , Vía de Señalización Wnt , Diferenciación Celular/fisiología , Humanos , Páncreas/citología , Células Madre/citología
20.
BMC Neurosci ; 9: 42, 2008 Apr 28.
Artículo en Inglés | MEDLINE | ID: mdl-18442397

RESUMEN

BACKGROUND: DNA-protein interactions in mature brain are increasingly recognized as key regulators for behavioral plasticity and neuronal dysfunction in chronic neuropsychiatric disease. However, chromatin assays typically lack single cell resolution, and therefore little is known about chromatin regulation of differentiated neuronal nuclei that reside in brain parenchyma intermingled with various types of non-neuronal cells. RESULTS: Here, we describe a protocol to selectively tag neuronal nuclei from adult brain - either by (anti-NeuN) immunolabeling or transgene-derived histone H2B-GFP fusion protein - for subsequent fluorescence-activated sorting and chromatin immunoprecipitation (ChIP). To illustrate an example, we compared histone H3 lysine 4 and 9 methylation marks at select gene promoters in neuronal, non-neuronal and unsorted chromatin from mouse forebrain and human cerebral cortex, and provide evidence for neuron-specific histone methylation signatures. CONCLUSION: With the modifications detailed in this protocol, the method can be used to collect nuclei from specific subtypes of neurons from any brain region for subsequent ChIP with native/un-fixed or crosslinked chromatin preparations. Starting with the harvest of brain tissue, ChIP-ready neuronal nuclei can be obtained within one day.


Asunto(s)
Encéfalo/metabolismo , Inmunoprecipitación de Cromatina/métodos , Cromatina/aislamiento & purificación , Biología Molecular/métodos , Neuroquímica/métodos , Neuronas/química , Adolescente , Adulto , Anciano , Animales , Antígenos Nucleares/análisis , Antígenos Nucleares/metabolismo , Encéfalo/citología , Núcleo Celular/química , Núcleo Celular/metabolismo , Niño , Cromatina/genética , Metilación de ADN , Citometría de Flujo , Histonas/análisis , Histonas/química , Histonas/metabolismo , Humanos , Ratones , Persona de Mediana Edad , Proteínas del Tejido Nervioso/análisis , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA