Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 296
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 176(5): 1098-1112.e18, 2019 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-30794774

RESUMEN

Increased levels of intestinal bile acids (BAs) are a risk factor for colorectal cancer (CRC). Here, we show that the convergence of dietary factors (high-fat diet) and dysregulated WNT signaling (APC mutation) alters BA profiles to drive malignant transformations in Lgr5-expressing (Lgr5+) cancer stem cells and promote an adenoma-to-adenocarcinoma progression. Mechanistically, we show that BAs that antagonize intestinal farnesoid X receptor (FXR) function, including tauro-ß-muricholic acid (T-ßMCA) and deoxycholic acid (DCA), induce proliferation and DNA damage in Lgr5+ cells. Conversely, selective activation of intestinal FXR can restrict abnormal Lgr5+ cell growth and curtail CRC progression. This unexpected role for FXR in coordinating intestinal self-renewal with BA levels implicates FXR as a potential therapeutic target for CRC.


Asunto(s)
Neoplasias Intestinales/metabolismo , Células Madre Neoplásicas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Animales , Ácidos y Sales Biliares/metabolismo , Línea Celular , Proliferación Celular/genética , Neoplasias Colorrectales/metabolismo , Ácido Desoxicólico/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Neoplasias Intestinales/genética , Intestinos , Hígado , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/fisiología , Organoides/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Factores de Riesgo , Transducción de Señal , Ácido Taurocólico/análogos & derivados , Ácido Taurocólico/metabolismo , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología
2.
Cell ; 173(5): 1135-1149.e15, 2018 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-29754817

RESUMEN

A primary cause of disease progression in type 2 diabetes (T2D) is ß cell dysfunction due to inflammatory stress and insulin resistance. However, preventing ß cell exhaustion under diabetic conditions is a major therapeutic challenge. Here, we identify the vitamin D receptor (VDR) as a key modulator of inflammation and ß cell survival. Alternative recognition of an acetylated lysine in VDR by bromodomain proteins BRD7 and BRD9 directs association to PBAF and BAF chromatin remodeling complexes, respectively. Mechanistically, ligand promotes VDR association with PBAF to effect genome-wide changes in chromatin accessibility and enhancer landscape, resulting in an anti-inflammatory response. Importantly, pharmacological inhibition of BRD9 promotes PBAF-VDR association to restore ß cell function and ameliorate hyperglycemia in murine T2D models. These studies reveal an unrecognized VDR-dependent transcriptional program underpinning ß cell survival and identifies the VDR:PBAF/BAF association as a potential therapeutic target for T2D.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , Células Secretoras de Insulina/efectos de los fármacos , Receptores de Calcitriol/metabolismo , Factores de Transcripción/metabolismo , Vitamina D/farmacología , Animales , Calcitriol/análogos & derivados , Calcitriol/farmacología , Ensamble y Desensamble de Cromatina , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Humanos , Insulina/sangre , Insulina/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Mutagénesis Sitio-Dirigida , Fosforilación Oxidativa/efectos de los fármacos , Unión Proteica , Interferencia de ARN , ARN Guía de Kinetoplastida/genética , ARN Interferente Pequeño/metabolismo , Receptores de Calcitriol/antagonistas & inhibidores , Receptores de Calcitriol/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Transcripción Genética/efectos de los fármacos
3.
Cell ; 165(7): 1644-1657, 2016 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-27238018

RESUMEN

Defects in circadian rhythm influence physiology and behavior with implications for the treatment of sleep disorders, metabolic disease, and cancer. Although core regulatory components of clock rhythmicity have been defined, insight into the mechanisms underpinning amplitude is limited. Here, we show that REV-ERBα, a core inhibitory component of clock transcription, is targeted for ubiquitination and subsequent degradation by the F-box protein FBXW7. By relieving REV-ERBα-dependent repression, FBXW7 provides an unrecognized mechanism for enhancing the amplitude of clock gene transcription. Cyclin-dependent kinase 1 (CDK1)-mediated phosphorylation of REV-ERBα is necessary for FBXW7 recognition. Moreover, targeted hepatic disruption of FBXW7 alters circadian expression of core clock genes and perturbs whole-body lipid and glucose levels. This CDK1-FBXW7 pathway controlling REV-ERBα repression defines an unexpected molecular mechanism for re-engaging the positive transcriptional arm of the clock, as well as a potential route to manipulate clock amplitude via small molecule CDK1 inhibition.


Asunto(s)
Ritmo Circadiano , Proteínas F-Box/metabolismo , Hígado/metabolismo , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , Línea Celular Tumoral , Relojes Circadianos , Proteínas F-Box/genética , Proteína 7 que Contiene Repeticiones F-Box-WD , Técnicas de Inactivación de Genes , Humanos , Metabolismo de los Lípidos , Ratones , Fosforilación , Procesamiento Proteico-Postraduccional , Transcriptoma , Ubiquitina-Proteína Ligasas/genética
4.
Mol Cell ; 83(19): 3421-3437.e11, 2023 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-37751740

RESUMEN

The nuclear receptor co-repressor (NCoR) complex mediates transcriptional repression dependent on histone deacetylation by histone deacetylase 3 (HDAC3) as a component of the complex. Unexpectedly, we found that signaling by the receptor activator of nuclear factor κB (RANK) converts the NCoR/HDAC3 co-repressor complex to a co-activator of AP-1 and NF-κB target genes that are required for mouse osteoclast differentiation. Accordingly, the dominant function of NCoR/HDAC3 complexes in response to RANK signaling is to activate, rather than repress, gene expression. Mechanistically, RANK signaling promotes RNA-dependent interaction of the transcriptional co-activator PGC1ß with the NCoR/HDAC3 complex, resulting in the activation of PGC1ß and inhibition of HDAC3 activity for acetylated histone H3. Non-coding RNAs Dancr and Rnu12, which are associated with altered human bone homeostasis, promote NCoR/HDAC3 complex assembly and are necessary for RANKL-induced osteoclast differentiation in vitro. These findings may be prototypic for signal-dependent functions of NCoR in other biological contexts.


Asunto(s)
Osteoclastos , ARN , Humanos , Ratones , Animales , Proteínas Co-Represoras/genética , Osteoclastos/metabolismo , Ligando RANK/genética , Co-Represor 1 de Receptor Nuclear/genética , Co-Represor 1 de Receptor Nuclear/metabolismo , Expresión Génica
5.
Immunity ; 54(7): 1561-1577.e7, 2021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34102100

RESUMEN

A common metabolic alteration in the tumor microenvironment (TME) is lipid accumulation, a feature associated with immune dysfunction. Here, we examined how CD8+ tumor infiltrating lymphocytes (TILs) respond to lipids within the TME. We found elevated concentrations of several classes of lipids in the TME and accumulation of these in CD8+ TILs. Lipid accumulation was associated with increased expression of CD36, a scavenger receptor for oxidized lipids, on CD8+ TILs, which also correlated with progressive T cell dysfunction. Cd36-/- T cells retained effector functions in the TME, as compared to WT counterparts. Mechanistically, CD36 promoted uptake of oxidized low-density lipoproteins (OxLDL) into T cells, and this induced lipid peroxidation and downstream activation of p38 kinase. Inhibition of p38 restored effector T cell functions in vitro, and resolution of lipid peroxidation by overexpression of glutathione peroxidase 4 restored functionalities in CD8+ TILs in vivo. Thus, an oxidized lipid-CD36 axis promotes intratumoral CD8+ T cell dysfunction and serves as a therapeutic avenue for immunotherapies.


Asunto(s)
Antígenos CD36/metabolismo , Linfocitos T CD8-positivos/metabolismo , Peroxidación de Lípido/fisiología , Lipoproteínas LDL/metabolismo , Neoplasias/metabolismo , Receptores Depuradores/metabolismo , Animales , Transporte Biológico/fisiología , Línea Celular Tumoral , Células HEK293 , Humanos , Leucocitos Mononucleares/metabolismo , Linfocitos Infiltrantes de Tumor/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microambiente Tumoral/fisiología
6.
Cell ; 161(5): 962-963, 2015 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-26000475

RESUMEN

How do cells maintain the fluidity of cellular membrane in response to temperature fluctuation? In this issue of Cell, Ma et al. identify a regulatory circuit involving a heat-induced acyl-CoA dehydrogenase that controls the lipid saturation level and the fluidity of cellular membranes by transcriptionally regulating a lipid desaturase.


Asunto(s)
Acil-CoA Deshidrogenasa/metabolismo , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/fisiología , Ácidos Grasos/metabolismo , Animales
7.
Immunity ; 52(6): 1057-1074.e7, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32362324

RESUMEN

Tissue-resident and recruited macrophages contribute to both host defense and pathology. Multiple macrophage phenotypes are represented in diseased tissues, but we lack deep understanding of mechanisms controlling diversification. Here, we investigate origins and epigenetic trajectories of hepatic macrophages during diet-induced non-alcoholic steatohepatitis (NASH). The NASH diet induced significant changes in Kupffer cell enhancers and gene expression, resulting in partial loss of Kupffer cell identity, induction of Trem2 and Cd9 expression, and cell death. Kupffer cell loss was compensated by gain of adjacent monocyte-derived macrophages that exhibited convergent epigenomes, transcriptomes, and functions. NASH-induced changes in Kupffer cell enhancers were driven by AP-1 and EGR that reprogrammed LXR functions required for Kupffer cell identity and survival to instead drive a scar-associated macrophage phenotype. These findings reveal mechanisms by which disease-associated environmental signals instruct resident and recruited macrophages to acquire distinct gene expression programs and corresponding functions.


Asunto(s)
Microambiente Celular/genética , Reprogramación Celular/genética , Epigénesis Genética , Regulación de la Expresión Génica , Células Mieloides/metabolismo , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Biomarcadores , Secuenciación de Inmunoprecipitación de Cromatina , Dieta , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Ontología de Genes , Secuenciación de Nucleótidos de Alto Rendimiento , Macrófagos del Hígado/inmunología , Macrófagos del Hígado/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Enfermedad del Hígado Graso no Alcohólico/patología , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Unión Proteica , Transducción de Señal , Análisis de la Célula Individual
8.
Cell ; 157(1): 255-66, 2014 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-24679540

RESUMEN

Isolation of genes encoding the receptors for steroids, retinoids, vitamin D, and thyroid hormone and their structural and functional analysis revealed an evolutionarily conserved template for nuclear hormone receptors. This discovery sparked identification of numerous genes encoding related proteins, termed orphan receptors. Characterization of these orphan receptors and, in particular, of the retinoid X receptor (RXR) positioned nuclear receptors at the epicenter of the "Big Bang" of molecular endocrinology. This Review provides a personal perspective on nuclear receptors and explores their integrated and coordinated signaling networks that are essential for multicellular life, highlighting the RXR heterodimer and its associated ligands and transcriptional mechanism.


Asunto(s)
Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores X Retinoide/metabolismo , Transducción de Señal , Animales , Endocrinología , Regulación de la Expresión Génica , Humanos , Receptores Citoplasmáticos y Nucleares/química , Receptores X Retinoide/química , Transcripción Genética
9.
Cell ; 159(1): 80-93, 2014 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-25259922

RESUMEN

The poor clinical outcome in pancreatic ductal adenocarcinoma (PDA) is attributed to intrinsic chemoresistance and a growth-permissive tumor microenvironment. Conversion of quiescent to activated pancreatic stellate cells (PSCs) drives the severe stromal reaction that characterizes PDA. Here, we reveal that the vitamin D receptor (VDR) is expressed in stroma from human pancreatic tumors and that treatment with the VDR ligand calcipotriol markedly reduced markers of inflammation and fibrosis in pancreatitis and human tumor stroma. We show that VDR acts as a master transcriptional regulator of PSCs to reprise the quiescent state, resulting in induced stromal remodeling, increased intratumoral gemcitabine, reduced tumor volume, and a 57% increase in survival compared to chemotherapy alone. This work describes a molecular strategy through which transcriptional reprogramming of tumor stroma enables chemotherapeutic response and suggests vitamin D priming as an adjunct in PDA therapy. PAPERFLICK:


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Calcitriol/análogos & derivados , Carcinoma Ductal Pancreático/tratamiento farmacológico , Neoplasias Pancreáticas/tratamiento farmacológico , Receptores de Calcitriol/metabolismo , Adenocarcinoma/patología , Animales , Calcitriol/farmacología , Carcinoma Ductal Pancreático/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Humanos , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Neoplasias Pancreáticas/patología , Pancreatitis/tratamiento farmacológico , Pancreatitis/prevención & control , Transducción de Señal , Células del Estroma/patología
10.
Cell ; 152(3): 381-2, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23374332

RESUMEN

Central circadian clock oscillators, located in the suprachiasmatic nucleus (SCN) of the hypothalamus, align peripheral clock systems with geosynchronous time. Gerber et al. (2013) identify actin polymerization and serum response factor (SRF) activation as key steps linking the central master clock to peripheral oscillators.

11.
Cell ; 153(3): 601-13, 2013 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-23622244

RESUMEN

Liver fibrosis is a reversible wound-healing response involving TGFß1/SMAD activation of hepatic stellate cells (HSCs). It results from excessive deposition of extracellular matrix components and can lead to impairment of liver function. Here, we show that vitamin D receptor (VDR) ligands inhibit HSC activation by TGFß1 and abrogate liver fibrosis, whereas Vdr knockout mice spontaneously develop hepatic fibrosis. Mechanistically, we show that TGFß1 signaling causes a redistribution of genome-wide VDR-binding sites (VDR cistrome) in HSCs and facilitates VDR binding at SMAD3 profibrotic target genes via TGFß1-dependent chromatin remodeling. In the presence of VDR ligands, VDR binding to the coregulated genes reduces SMAD3 occupancy at these sites, inhibiting fibrosis. These results reveal an intersecting VDR/SMAD genomic circuit that regulates hepatic fibrogenesis and define a role for VDR as an endocrine checkpoint to modulate the wound-healing response in liver. Furthermore, the findings suggest VDR ligands as a potential therapy for liver fibrosis.


Asunto(s)
Redes Reguladoras de Genes , Hígado/metabolismo , Hígado/patología , Receptores de Calcitriol/metabolismo , Transducción de Señal , Animales , Calcitriol/análogos & derivados , Fibrosis/prevención & control , Estudio de Asociación del Genoma Completo , Células Estrelladas Hepáticas , Hígado/lesiones , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratas , Receptores de Calcitriol/agonistas , Proteína smad3/metabolismo , Transcriptoma , Factor de Crecimiento Transformador beta1/metabolismo
12.
Nature ; 604(7905): 337-342, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35355021

RESUMEN

Decades of work have elucidated cytokine signalling and transcriptional pathways that control T cell differentiation and have led the way to targeted biologic therapies that are effective in a range of autoimmune, allergic and inflammatory diseases. Recent evidence indicates that obesity and metabolic disease can also influence the immune system1-7, although the mechanisms and effects on immunotherapy outcomes remain largely unknown. Here, using two models of atopic dermatitis, we show that lean and obese mice mount markedly different immune responses. Obesity converted the classical type 2 T helper (TH2)-predominant disease associated with atopic dermatitis to a more severe disease with prominent TH17 inflammation. We also observed divergent responses to biologic therapies targeting TH2 cytokines, which robustly protected lean mice but exacerbated disease in obese mice. Single-cell RNA sequencing coupled with genome-wide binding analyses revealed decreased activity of nuclear receptor peroxisome proliferator-activated receptor-γ (PPARγ) in TH2 cells from obese mice relative to lean mice. Conditional ablation of PPARγ in T cells revealed that PPARγ is required to focus the in vivo TH response towards a TH2-predominant state and prevent aberrant non-TH2 inflammation. Treatment of obese mice with a small-molecule PPARγ agonist limited development of TH17 pathology and unlocked therapeutic responsiveness to targeted anti-TH2 biologic therapies. These studies reveal the effects of obesity on immunological disease and suggest a precision medicine approach to target the immune dysregulation caused by obesity.


Asunto(s)
Dermatitis Atópica , PPAR gamma , Animales , Citocinas/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Obesidad/metabolismo , PPAR gamma/agonistas , PPAR gamma/metabolismo , Medicina de Precisión , Análisis de Secuencia de ARN , Células Th2/metabolismo
13.
Nature ; 599(7884): 296-301, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34707293

RESUMEN

Adipocytes increase energy expenditure in response to prolonged sympathetic activation via persistent expression of uncoupling protein 1 (UCP1)1,2. Here we report that the regulation of glycogen metabolism by catecholamines is critical for UCP1 expression. Chronic ß-adrenergic activation leads to increased glycogen accumulation in adipocytes expressing UCP1. Adipocyte-specific deletion of a scaffolding protein, protein targeting to glycogen (PTG), reduces glycogen levels in beige adipocytes, attenuating UCP1 expression and responsiveness to cold or ß-adrenergic receptor-stimulated weight loss in obese mice. Unexpectedly, we observed that glycogen synthesis and degradation are increased in response to catecholamines, and that glycogen turnover is required to produce reactive oxygen species leading to the activation of p38 MAPK, which drives UCP1 expression. Thus, glycogen has a key regulatory role in adipocytes, linking glucose metabolism to thermogenesis.


Asunto(s)
Adipocitos/metabolismo , Glucosa/metabolismo , Glucógeno/metabolismo , Homeostasis , Termogénesis , Adaptación Fisiológica , Adipocitos Beige/metabolismo , Animales , Frío , Metabolismo Energético , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones , Ratones Noqueados , Proteína Desacopladora 1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
14.
Proc Natl Acad Sci U S A ; 121(9): e2320129121, 2024 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-38377195

RESUMEN

Despite numerous female contraceptive options, nearly half of all pregnancies are unintended. Family planning choices for men are currently limited to unreliable condoms and invasive vasectomies with questionable reversibility. Here, we report the development of an oral contraceptive approach based on transcriptional disruption of cyclical gene expression patterns during spermatogenesis. Spermatogenesis involves a continuous series of self-renewal and differentiation programs of spermatogonial stem cells (SSCs) that is regulated by retinoic acid (RA)-dependent activation of receptors (RARs), which control target gene expression through association with corepressor proteins. We have found that the interaction between RAR and the corepressor silencing mediator of retinoid and thyroid hormone receptors (SMRT) is essential for spermatogenesis. In a genetically engineered mouse model that negates SMRT-RAR binding (SMRTmRID mice), the synchronized, cyclic expression of RAR-dependent genes along the seminiferous tubules is disrupted. Notably, the presence of an RA-resistant SSC population that survives RAR de-repression suggests that the infertility attributed to the loss of SMRT-mediated repression is reversible. Supporting this notion, we show that inhibiting the action of the SMRT complex with chronic, low-dose oral administration of a histone deacetylase inhibitor reversibly blocks spermatogenesis and fertility without affecting libido. This demonstration validates pharmacologic targeting of the SMRT repressor complex for non-hormonal male contraception.


Asunto(s)
Proteínas de Unión al ADN , Proteínas Represoras , Humanos , Femenino , Masculino , Animales , Ratones , Proteínas de Unión al ADN/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Proteínas Co-Represoras/genética , Co-Represor 2 de Receptor Nuclear/genética , Tretinoina/farmacología , Anticoncepción , Co-Represor 1 de Receptor Nuclear
15.
Proc Natl Acad Sci U S A ; 121(2): e2316104121, 2024 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-38165941

RESUMEN

The nuclear receptor corepressor (NCoR) forms a complex with histone deacetylase 3 (HDAC3) that mediates repressive functions of unliganded nuclear receptors and other transcriptional repressors by deacetylation of histone substrates. Recent studies provide evidence that NCoR/HDAC3 complexes can also exert coactivator functions in brown adipocytes by deacetylating and activating PPARγ coactivator 1α (PGC1α) and that signaling via receptor activator of nuclear factor kappa-B (RANK) promotes the formation of a stable NCoR/HDAC3/PGC1ß complex that coactivates nuclear factor kappa-B (NFκB)- and activator protein 1 (AP-1)-dependent genes required for osteoclast differentiation. Here, we demonstrate that activation of Toll-like receptor (TLR) 4, but not TLR3, the interleukin 4 (IL4) receptor nor the Type I interferon receptor, also promotes assembly of an NCoR/HDAC3/PGC1ß coactivator complex. Receptor-specific utilization of TNF receptor-associated factor 6 (TRAF6) and downstream activation of extracellular signal-regulated kinase 1 (ERK1) and TANK-binding kinase 1 (TBK1) accounts for the common ability of RANK and TLR4 to drive assembly of an NCoR/HDAC3/PGC1ß complex in macrophages. ERK1, the p65 component of NFκB, and the p300 histone acetyltransferase (HAT) are also components of the induced complex and are associated with local histone acetylation and transcriptional activation of TLR4-dependent enhancers and promoters. These observations identify a TLR4/TRAF6-dependent signaling pathway that converts NCoR from a corepressor of nuclear receptors to a coactivator of NFκB and AP-1 that may be relevant to functions of NCoR in other developmental and homeostatic processes.


Asunto(s)
Histonas , Factor 6 Asociado a Receptor de TNF , Activación Transcripcional , Proteínas Co-Represoras/genética , Histonas/genética , Histonas/metabolismo , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Factor de Transcripción AP-1/metabolismo , Receptor Toll-Like 4/metabolismo , Transducción de Señal , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo
16.
Cell ; 145(4): 607-21, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565617

RESUMEN

Class IIa histone deacetylases (HDACs) are signal-dependent modulators of transcription with established roles in muscle differentiation and neuronal survival. We show here that in liver, class IIa HDACs (HDAC4, 5, and 7) are phosphorylated and excluded from the nucleus by AMPK family kinases. In response to the fasting hormone glucagon, class IIa HDACs are rapidly dephosphorylated and translocated to the nucleus where they associate with the promoters of gluconeogenic enzymes such as G6Pase. In turn, HDAC4/5 recruit HDAC3, which results in the acute transcriptional induction of these genes via deacetylation and activation of FOXO family transcription factors. Loss of class IIa HDACs in murine liver results in inhibition of FOXO target genes and lowers blood glucose, resulting in increased glycogen storage. Finally, suppression of class IIa HDACs in mouse models of type 2 diabetes ameliorates hyperglycemia, suggesting that inhibitors of class I/II HDACs may be potential therapeutics for metabolic syndrome.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Glucosa/metabolismo , Histona Desacetilasas/metabolismo , Proteínas Quinasas Activadas por AMP , Acetilación , Animales , Núcleo Celular/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Proteína Forkhead Box O1 , Glucagón/metabolismo , Gluconeogénesis , Homeostasis , Ratones , Fosforilación , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal
17.
Cell ; 147(4): 827-39, 2011 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-22078881

RESUMEN

Transcriptional coregulators control the activity of many transcription factors and are thought to have wide-ranging effects on gene expression patterns. We show here that muscle-specific loss of nuclear receptor corepressor 1 (NCoR1) in mice leads to enhanced exercise endurance due to an increase of both muscle mass and of mitochondrial number and activity. The activation of selected transcription factors that control muscle function, such as MEF2, PPARß/δ, and ERRs, underpins these phenotypic alterations. NCoR1 levels are decreased in conditions that require fat oxidation, resetting transcriptional programs to boost oxidative metabolism. Knockdown of gei-8, the sole C. elegans NCoR homolog, also robustly increased muscle mitochondria and respiration, suggesting conservation of NCoR1 function. Collectively, our data suggest that NCoR1 plays an adaptive role in muscle physiology and that interference with NCoR1 action could be used to improve muscle function.


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Músculo Esquelético/metabolismo , Co-Represor 1 de Receptor Nuclear/metabolismo , Animales , Proteínas de Caenorhabditis elegans/genética , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Mitocondrias Musculares/metabolismo , Desarrollo de Músculos , Co-Represor 1 de Receptor Nuclear/genética , PPAR delta/metabolismo , PPAR-beta/metabolismo , Condicionamiento Físico Animal
18.
Nature ; 586(7830): 606-611, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32814902

RESUMEN

Islets derived from stem cells hold promise as a therapy for insulin-dependent diabetes, but there remain challenges towards achieving this goal1-6. Here we generate human islet-like organoids (HILOs) from induced pluripotent stem cells and show that non-canonical WNT4 signalling drives the metabolic maturation necessary for robust ex vivo glucose-stimulated insulin secretion. These functionally mature HILOs contain endocrine-like cell types that, upon transplantation, rapidly re-establish glucose homeostasis in diabetic NOD/SCID mice. Overexpression of the immune checkpoint protein programmed death-ligand 1 (PD-L1) protected HILO xenografts such that they were able to restore glucose homeostasis in immune-competent diabetic mice for 50 days. Furthermore, ex vivo stimulation with interferon-γ induced endogenous PD-L1 expression and restricted T cell activation and graft rejection. The generation of glucose-responsive islet-like organoids that are able to avoid immune detection provides a promising alternative to cadaveric and device-dependent therapies in the treatment of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Evasión Inmune , Islotes Pancreáticos/citología , Islotes Pancreáticos/inmunología , Organoides/citología , Organoides/inmunología , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Línea Celular , Epigénesis Genética , Femenino , Glucosa/metabolismo , Rechazo de Injerto , Xenoinjertos , Homeostasis , Humanos , Tolerancia Inmunológica , Secreción de Insulina , Trasplante de Islotes Pancreáticos , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Organoides/trasplante , Linfocitos T/citología , Linfocitos T/inmunología , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt4/metabolismo , Proteína Wnt4/farmacología
19.
Proc Natl Acad Sci U S A ; 120(33): e2300036120, 2023 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-37549292

RESUMEN

While the world is rapidly transforming into a superaging society, pharmaceutical approaches to treat sarcopenia have hitherto not been successful due to their insufficient efficacy and failure to specifically target skeletal muscle cells (skMCs). Although electrical stimulation (ES) is emerging as an alternative intervention, its efficacy toward treating sarcopenia remains unexplored. In this study, we demonstrate a silver electroceutical technology with the potential to treat sarcopenia. First, we developed a high-throughput ES screening platform that can simultaneously stimulate 15 independent conditions, while utilizing only a small number of human-derived primary aged/young skMCs (hAskMC/hYskMC). The in vitro screening showed that specific ES conditions induced hypertrophy and rejuvenation in hAskMCs, and the optimal ES frequency in hAskMCs was different from that in hYskMCs. When applied to aged mice in vivo, specific ES conditions improved the prevalence and thickness of Type IIA fibers, along with biomechanical attributes, toward a younger skMC phenotype. This study is expected to pave the way toward an electroceutical treatment for sarcopenia with minimal side effects and help realize personalized bioelectronic medicine.


Asunto(s)
Sarcopenia , Animales , Humanos , Ratones , Fibras Musculares Esqueléticas , Músculo Esquelético/fisiología , Fenotipo , Sarcopenia/terapia , Plata
20.
Proc Natl Acad Sci U S A ; 120(21): e2217826120, 2023 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-37192160

RESUMEN

Molecular classification of gastric cancer (GC) identified a subgroup of patients showing chemoresistance and poor prognosis, termed SEM (Stem-like/Epithelial-to-mesenchymal transition/Mesenchymal) type in this study. Here, we show that SEM-type GC exhibits a distinct metabolic profile characterized by high glutaminase (GLS) levels. Unexpectedly, SEM-type GC cells are resistant to glutaminolysis inhibition. We show that under glutamine starvation, SEM-type GC cells up-regulate the 3 phosphoglycerate dehydrogenase (PHGDH)-mediated mitochondrial folate cycle pathway to produce NADPH as a reactive oxygen species scavenger for survival. This metabolic plasticity is associated with globally open chromatin structure in SEM-type GC cells, with ATF4/CEBPB identified as transcriptional drivers of the PHGDH-driven salvage pathway. Single-nucleus transcriptome analysis of patient-derived SEM-type GC organoids revealed intratumoral heterogeneity, with stemness-high subpopulations displaying high GLS expression, a resistance to GLS inhibition, and ATF4/CEBPB activation. Notably, coinhibition of GLS and PHGDH successfully eliminated stemness-high cancer cells. Together, these results provide insight into the metabolic plasticity of aggressive GC cells and suggest a treatment strategy for chemoresistant GC patients.


Asunto(s)
Fosfoglicerato-Deshidrogenasa , Neoplasias Gástricas , Humanos , Fosfoglicerato-Deshidrogenasa/genética , Fosfoglicerato-Deshidrogenasa/metabolismo , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/genética , Línea Celular Tumoral , Glutamina/metabolismo , Nutrientes
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA