Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Development ; 149(7)2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35297993

RESUMEN

Beige adipocytes have a discrete developmental origin and possess notable plasticity in their thermogenic capacity in response to various environmental cues, but the transcriptional machinery controlling beige adipocyte development and thermogenesis remains largely unknown. By analyzing beige adipocyte-specific knockout mice, we identified a transcription factor, forkhead box P4 (FOXP4), that differentially governs beige adipocyte differentiation and activation. Depletion of Foxp4 in progenitor cells impaired beige cell early differentiation. However, we observed that ablation of Foxp4 in differentiated adipocytes profoundly potentiated their thermogenesis capacity upon cold exposure. Of note, the outcome of Foxp4 deficiency on UCP1-mediated thermogenesis was confined to beige adipocytes, rather than to brown adipocytes. Taken together, we suggest that FOXP4 primes beige adipocyte early differentiation, but attenuates their activation by potent transcriptional repression of the thermogenic program.


Asunto(s)
Adipocitos Beige , Adipocitos Marrones , Animales , Diferenciación Celular/genética , Regulación de la Expresión Génica , Ratones , Termogénesis/genética
2.
Immunity ; 39(2): 272-85, 2013 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-23973223

RESUMEN

Regulatory T (Treg) cells suppress inflammatory immune responses and autoimmunity caused by self-reactive T cells. The key Treg cell transcription factor Foxp3 is downregulated during inflammation to allow for the acquisition of effector T cell-like functions. Here, we demonstrate that stress signals elicited by proinflammatory cytokines and lipopolysaccharides lead to the degradation of Foxp3 through the action of the E3 ubiquitin ligase Stub1. Stub1 interacted with Foxp3 to promote its K48-linked polyubiquitination in an Hsp70-dependent manner. Knockdown of endogenous Stub1 or Hsp70 prevented Foxp3 degradation. Furthermore, the overexpression of Stub1 in Treg cells abrogated their ability to suppress inflammatory immune responses in vitro and in vivo and conferred a T-helper-1-cell-like phenotype. Our results demonstrate the critical role of the stress-activated Stub1-Hsp70 complex in promoting Treg cell inactivation, thus providing a potential therapeutic target for the intervention against autoimmune disease, infection, and cancer.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Proteínas HSP70 de Choque Térmico/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Células Cultivadas , Citocinas/metabolismo , Inhibidores Enzimáticos , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Humanos , Imidazoles , Inflamación/genética , Inflamación/inmunología , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos BALB C , Fenotipo , Piridinas , Interferencia de ARN , ARN Interferente Pequeño , Linfocitos T Colaboradores-Inductores/inmunología , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
3.
Proc Natl Acad Sci U S A ; 115(35): 8799-8804, 2018 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-30104377

RESUMEN

Fundamental human traits, such as language and bipedalism, are associated with a range of anatomical adaptations in craniofacial shaping and skeletal remodeling. However, it is unclear how such morphological features arose during hominin evolution. FOXP2 is a brain-expressed transcription factor implicated in a rare disorder involving speech apraxia and language impairments. Analysis of its evolutionary history suggests that this gene may have contributed to the emergence of proficient spoken language. In the present study, through analyses of skeleton-specific knockout mice, we identified roles of Foxp2 in skull shaping and bone remodeling. Selective ablation of Foxp2 in cartilage disrupted pup vocalizations in a similar way to that of global Foxp2 mutants, which may be due to pleiotropic effects on craniofacial morphogenesis. Our findings also indicate that Foxp2 helps to regulate strength and length of hind limbs and maintenance of joint cartilage and intervertebral discs, which are all anatomical features that are susceptible to adaptations for bipedal locomotion. In light of the known roles of Foxp2 in brain circuits that are important for motor skills and spoken language, we suggest that this gene may have been well placed to contribute to coevolution of neural and anatomical adaptations related to speech and bipedal locomotion.


Asunto(s)
Remodelación Ósea/genética , Factores de Transcripción Forkhead , Locomoción/genética , Mutación , Proteínas Represoras , Cráneo/metabolismo , Vocalización Animal , Animales , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Miembro Posterior/metabolismo , Humanos , Ratones , Ratones Noqueados , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
5.
Immunity ; 30(1): 155-67, 2009 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-19144320

RESUMEN

Multipotential naive CD4(+) T cells differentiate into distinct lineages including T helper 1 (Th1), Th2, Th17, and inducible T regulatory (iTreg) cells. The remarkable diversity of CD4(+) T cells begs the question whether the observed changes reflect terminal differentiation with heritable epigenetic modifications or plasticity in T cell responses. We generated genome-wide histone H3 lysine 4 (H3K4) and lysine 27 (H3K27) trimethylation maps in naive, Th1, Th2, Th17, iTreg, and natural Treg (nTreg) cells. We found that although modifications of signature-cytokine genes (Ifng, Il4, and Il17) partially conform to the expectation of lineage commitment, genes encoding transcription factors like Tbx21 exhibit a broad spectrum of epigenetic states, consistent with our demonstration of T-bet and interferon-gamma induction in nTreg cells. Our data suggest an epigenetic mechanism underlying the specificity and plasticity of effector and regulatory T cells and also provide a framework for understanding complexity of CD4(+) T helper cell differentiation.


Asunto(s)
Antígenos CD4/metabolismo , Linfocitos T CD4-Positivos/inmunología , Mapeo Cromosómico , Histonas/metabolismo , Oxidorreductasas N-Desmetilantes/genética , Subgrupos de Linfocitos T/inmunología , Animales , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Proteínas de Unión al ADN , Histona Demetilasas con Dominio de Jumonji , Ratones , Ratones Endogámicos C57BL , Oxidorreductasas N-Desmetilantes/inmunología , Modificación Traduccional de las Proteínas , Proteína 2 de Unión a Retinoblastoma
6.
Dev Biol ; 398(2): 242-54, 2015 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-25527076

RESUMEN

Osteoblast induction and differentiation in developing long bones is dynamically controlled by the opposing action of transcriptional activators and repressors. In contrast to the long list of activators that have been discovered over past decades, the network of repressors is not well-defined. Here we identify the expression of Foxp1/2/4 proteins, comprised of Forkhead-box (Fox) transcription factors of the Foxp subfamily, in both perichondrial skeletal progenitors and proliferating chondrocytes during endochondral ossification. Mice carrying loss-of-function and gain-of-function Foxp mutations had gross defects in appendicular skeleton formation. At the cellular level, over-expression of Foxp1/2/4 in chondroctyes abrogated osteoblast formation and chondrocyte hypertrophy. Conversely, single or compound deficiency of Foxp1/2/4 in skeletal progenitors or chondrocytes resulted in premature osteoblast differentiation in the perichondrium, coupled with impaired proliferation, survival, and hypertrophy of chondrocytes in the growth plate. Foxp1/2/4 and Runx2 proteins interacted in vitro and in vivo, and Foxp1/2/4 repressed Runx2 transactivation function in heterologous cells. This study establishes Foxp1/2/4 proteins as coordinators of osteogenesis and chondrocyte hypertrophy in developing long bones and suggests that a novel transcriptional repressor network involving Foxp1/2/4 may regulate Runx2 during endochondral ossification.


Asunto(s)
Condrocitos/metabolismo , Factores de Transcripción Forkhead/metabolismo , Osteogénesis , Proteínas Represoras/metabolismo , Animales , Huesos/metabolismo , Células COS , Calcificación Fisiológica , Chlorocebus aethiops , Condrocitos/patología , Condrogénesis/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Extremidades/embriología , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Células HEK293 , Humanos , Hipertrofia , Integrasas/metabolismo , Ratones Transgénicos , Unión Proteica , Proteínas Represoras/deficiencia , Proteínas Represoras/genética
7.
Eur J Immunol ; 45(9): 2650-60, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26173091

RESUMEN

Osteoblasts and perivascular stromal cells constitute essential niches for HSC self-renewal and maintenance in the bone marrow. Wnt signaling is important to maintain HSC integrity. However, the paracrine role of Wnt proteins in osteoblasts-supported HSC maintenance and differentiation remains unclear. Here, we investigated hematopoiesis in mice with Wntless (Wls) deficiency in osteoblasts or Nestin-positive mesenchymal progenitor cells, which presumptively block Wnt secretion in osteoblasts. We detected defective B-cell lymphopoiesis and abnormal T-cell infiltration in the bone marrow of Wls mutant mice. Notably, no impact on HSC frequency and repopulation in the bone marrow was observed with the loss of osteoblastic Wls. Our findings revealed a supportive role of Wnts in osteoblasts-regulated B-cell lymphopoiesis. They also suggest a preferential niche role of osteoblastic Wnts for lymphoid cells rather than HSCs, providing new clues for the molecular nature of distinct niches occupied by different hematopoietic cells.


Asunto(s)
Linfocitos B/inmunología , Hematopoyesis/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Linfopoyesis/genética , Receptores Acoplados a Proteínas G/genética , Nicho de Células Madre/inmunología , Linfocitos T/inmunología , Vía de Señalización Wnt , Animales , Linfocitos B/patología , Células de la Médula Ósea/citología , Células de la Médula Ósea/inmunología , Huesos/citología , Huesos/inmunología , Diferenciación Celular , Movimiento Celular , Regulación de la Expresión Génica , Hematopoyesis/inmunología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Péptidos y Proteínas de Señalización Intracelular/inmunología , Linfopoyesis/inmunología , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Noqueados , Nestina/genética , Nestina/inmunología , Osteoblastos/citología , Osteoblastos/inmunología , Comunicación Paracrina/genética , Comunicación Paracrina/inmunología , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/inmunología , Nicho de Células Madre/genética , Linfocitos T/patología
8.
Dev Biol ; 387(1): 64-72, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24394376

RESUMEN

Defects of the ventral body wall are prevalent birth anomalies marked by deficiencies in body wall closure, hypoplasia of the abdominal musculature and multiple malformations across a gamut of organs. However, the mechanisms underlying ventral body wall defects remain elusive. Here, we investigated the role of Wnt signaling in ventral body wall development by inactivating Wls or ß-catenin in murine abdominal ectoderm. The loss of Wls in the ventral epithelium, which blocks the secretion of Wnt proteins, resulted in dysgenesis of ventral musculature and genito-urinary tract during embryonic development. Molecular analyses revealed that the dermis and myogenic differentiation in the underlying mesenchymal progenitor cells was perturbed by the loss of ectodermal Wls. The activity of the Wnt-Pitx2 axis was impaired in the ventral mesenchyme of the mutant body wall, which partially accounted for the defects in ventral musculature formation. In contrast, epithelial depletion of ß-catenin or Wnt5a did not resemble the body wall defects in the ectodermal Wls mutant. These findings indicate that ectodermal Wnt signaling instructs the underlying mesodermal specification and abdominal musculature formation during ventral body wall development, adding evidence to the theory that ectoderm-mesenchyme signaling is a potential unifying mechanism for the origin of ventral body wall defects.


Asunto(s)
Abdomen/embriología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Desarrollo de Músculos/genética , Receptores Acoplados a Proteínas G/fisiología , Vía de Señalización Wnt/genética , beta Catenina/fisiología , Abdomen/crecimiento & desarrollo , Animales , Tipificación del Cuerpo/genética , Diferenciación Celular/genética , Ectodermo/embriología , Ectodermo/crecimiento & desarrollo , Ectodermo/metabolismo , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Mesodermo/embriología , Mesodermo/crecimiento & desarrollo , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores Acoplados a Proteínas G/genética , Factores de Transcripción/genética , Sistema Urogenital/embriología , Sistema Urogenital/crecimiento & desarrollo , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a , beta Catenina/genética , Proteína del Homeodomínio PITX2
9.
J Biol Chem ; 288(22): 15537-46, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23609452

RESUMEN

The abundant expression of IFNγ in Th-inducing POK (ThPOK)-deficient CD4(+) T cells requires the activation of Eomesodermin (Eomes); however, the underlying mechanism of this phenomenon remains unclear. Here we report that ThPOK binds directly to the promoter region of the Eomes gene to repress its expression in CD4(+) T cells. We identified the histone acetyltransferase TIP60 as a co-repressor of ThPOK-target genes, where ectopically expressed TIP60 increased ThPOK protein stability by promoting its acetylation at its Lys(360) residue to then augment the transcriptional repression of Eomes. Moreover, knockdown of endogenous TIP60 abolished the stabilization of ThPOK in CD4(+) T cells, which led to the transcriptional activation of Eomes and increased production of IFNγ. Our results reveal a novel pathway by which TIP60 and ThPOK synergistically suppresses Eomes function and IFNγ production, which could contribute to the regulation of inflammation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proteínas de Unión al ADN/metabolismo , Regulación de la Expresión Génica/inmunología , Histona Acetiltransferasas/metabolismo , Proteínas Represoras/metabolismo , Proteínas de Dominio T Box/metabolismo , Factores de Transcripción/metabolismo , Acetilación , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Regulación de la Expresión Génica/genética , Células HEK293 , Histona Acetiltransferasas/genética , Histona Acetiltransferasas/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/metabolismo , Interferón gamma/biosíntesis , Interferón gamma/genética , Interferón gamma/inmunología , Lisina Acetiltransferasa 5 , Regiones Promotoras Genéticas/genética , Regiones Promotoras Genéticas/inmunología , Estabilidad Proteica , Proteínas Represoras/genética , Proteínas Represoras/inmunología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología
10.
J Biol Chem ; 288(13): 9373-82, 2013 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-23395819

RESUMEN

The expression of the transcription factor GATA3 in FOXP3(+) regulatory T (Treg) cells is crucial for their physiological function in limiting inflammatory responses. Although other studies have shown how T cell receptor (TcR) signals induce the up-regulation of GATA3 expression in Treg cells, the underlying mechanism that maintains GATA3 expression in Treg cells remains unclear. Here, we show how USP21 interacts with and stabilizes GATA3 by mediating its deubiquitination. In a T cell line model, we found that TcR stimulation promoted USP21 expression, which was further up-regulated in the presence of FOXP3. The USP21 mutant C221A reduced its capacity to stabilize GATA3 expression, and its knockdown led to the down-regulation of GATA3 protein expression in Treg cells. Furthermore, we found that FOXP3 could directly bind to the USP21 gene promoter and activated its transcription upon TcR stimulation. Finally, USP21, GATA3, and FOXP3 were found up-regulated in Treg cells that were isolated from asthmatic subjects. In summary, we have identified a USP21-mediated pathway that promotes GATA3 stabilization and expression at the post-translational level. We propose that this pathway forms an important signaling loop that stabilizes the expression of GATA3 in Treg cells.


Asunto(s)
Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Ubiquitina Tiolesterasa/metabolismo , Adolescente , Adulto , Asma/metabolismo , Línea Celular Tumoral , Factores de Transcripción Forkhead/biosíntesis , Células HEK293 , Humanos , Activación de Linfocitos , Persona de Mediana Edad , Regiones Promotoras Genéticas , Procesamiento Proteico-Postraduccional , Linfocitos T/metabolismo
11.
Dev Biol ; 369(2): 308-18, 2012 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-22819676

RESUMEN

It is generally thought that vertebral patterning and identity are globally determined prior to somite formation. Relatively little is known about the regulators of vertebral specification after somite segmentation. Here, we demonstrated that Ndrg2, a tumor suppressor gene, was dynamically expressed in the presomitic mesoderm (PSM) and at early stage of differentiating somites. Loss of Ndrg2 in mice resulted in vertebral homeotic transformations in thoracic/lumbar and lumbar/sacral transitional regions in a dose-dependent manner. Interestingly, the inactivation of Ndrg2 in osteoblasts or chondrocytes caused defects resembling those observed in Ndrg2(-/-) mice, with a lower penetrance. In addition, forced overexpression of Ndrg2 in osteoblasts or chondrocytes also conferred vertebral defects, which were distinct from those in Ndrg2(-/-) mice. These genetic analyses revealed that Ndrg2 modulates vertebral identity in segmented somites rather than in the PSM. At the molecular level, combinatory alterations of the amount of Hoxc8-11 gene transcripts were detected in the differentiating somites of Ndrg2(-/-) embryos, which may partially account for the vertebral defects in Ndrg2 mutants. Nevertheless, Bmp/Smad signaling activity was elevated in the differentiating somites of Ndrg2(-/-) embryos. Collectively, our findings unveiled Ndrg2 as a novel regulator of vertebral specification in differentiating somites.


Asunto(s)
Proteínas/metabolismo , Somitos/embriología , Somitos/metabolismo , Columna Vertebral/embriología , Columna Vertebral/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Secuencia de Bases , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/fisiología , Proteínas Morfogenéticas Óseas/genética , Proteínas Morfogenéticas Óseas/metabolismo , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Genes Homeobox , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Mutantes , Ratones Transgénicos , Embarazo , Proteínas/genética , Transducción de Señal , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
12.
J Exp Med ; 204(1): 65-71, 2007 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-17227909

RESUMEN

Although required for many fundamental immune processes, ranging from self-tolerance to pathogen immunity, interleukin (IL)-2 production is transient, and the mechanisms underlying this brevity remain unclear. These studies reveal that helper T cell IL-2 production is limited by a classic negative feedback loop that functions autonomously or in collaboration with other common gamma chain (IL-4 and IL-7) and IL-6/IL-12 family cytokines (IL-12 and IL-27). Consistent with this model for cytokine-dependent regulation, they also demonstrate that the inhibitory effect can be mediated by several signal transducer and activator of transcription (STAT) family transcription factors, namely STAT5, STAT4, and STAT6. Collectively, these findings establish that IL-2 production is limited by a network of autocrine and paracrine signals that are readily available during acute inflammatory responses and, thus, provide a cellular and molecular basis for its transient pattern of expression.


Asunto(s)
Citocinas/metabolismo , Interleucina-2/biosíntesis , Factores de Transcripción STAT/metabolismo , Linfocitos T Colaboradores-Inductores/inmunología , Animales , Retroalimentación , Inmunización , Interleucina-2/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Ovalbúmina/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Factores de Transcripción STAT/deficiencia , Factores de Transcripción STAT/genética , Factor de Transcripción STAT4/deficiencia , Factor de Transcripción STAT4/genética , Factor de Transcripción STAT4/metabolismo , Factor de Transcripción STAT5/deficiencia , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/metabolismo , Factor de Transcripción STAT6/deficiencia , Factor de Transcripción STAT6/genética , Factor de Transcripción STAT6/metabolismo , Transducción de Señal , Células TH1/inmunología , Células Th2/inmunología
13.
Stem Cell Res Ther ; 14(1): 188, 2023 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-37507770

RESUMEN

BACKGROUND: Mesenchymal stem cells (MSCs) are widely used in a variety of tissue regeneration and clinical trials due to their multiple differentiation potency. However, it remains challenging to maintain their replicative capability during in vitro passaging while preventing their premature cellular senescence. Forkhead Box P1 (FOXP1), a FOX family transcription factor, has been revealed to regulate MSC cell fate commitment and self-renewal capacity in our previous study. METHODS: Mass spectra analysis was performed to identify acetylation sites in FOXP1 protein. Single and double knockout mice of FOXP1 and HDAC7 were generated and analyzed with bone marrow MSCs properties. Gene engineering in human embryonic stem cell (hESC)-derived MSCs was obtained to evaluate the impact of FOXP1 key modification on MSC self-renewal potency. RESULTS: FOXP1 is deacetylated and potentiated by histone deacetylase 7 (HDAC7) in MSCs. FOXP1 and HDAC7 cooperatively sustain bone marrow MSC self-renewal potency while attenuating their cellular senescence. A mutation within human FOXP1 at acetylation site (T176G) homologous to murine FOXP1 T172G profoundly augmented MSC expansion capacity during early passages. CONCLUSION: These findings reveal a heretofore unanticipated mechanism by which deacetylation of FOXP1 potentiates self-renewal of MSC and protects them from cellular senescence. Acetylation of FOXP1 residue T172 as a critical modification underlying MSC proliferative capacity. We suggest that in vivo gene editing of FOXP1 may provide a novel avenue for manipulating MSC capability during large-scale expansion in clinical trials.


Asunto(s)
Senescencia Celular , Células Madre Mesenquimatosas , Animales , Humanos , Ratones , Diferenciación Celular/genética , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Histona Desacetilasas/genética , Células Madre Mesenquimatosas/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo
14.
J Bone Miner Res ; 36(10): 2017-2026, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34131944

RESUMEN

Adiponectin (AdipoQ), a hormone abundantly secreted by adipose tissues, has multiple beneficial functions, including insulin sensitization as well as lipid and glucose metabolism. It has been reported that bone controls energy metabolism through an endocrine-based mechanism. In this study, we observed that bone also acts as an important endocrine source for AdipoQ, and its capacity in osteoblasts is controlled by the forkhead box P1 (FOXP1) transcriptional factor. Deletion of the Foxp1 gene in osteoblasts led to augmentation of AdipoQ levels accompanied by fueled energy expenditure in adipose tissues. In contrast, overexpression of Foxp1 in bones impaired AdipoQ secretion and restrained energy consumption. Chromatin immunoprecipitation sequencing (ChIP-seq) analysis revealed that AdipoQ expression, which increases as a function of bone age, is directly controlled by FOXP1. Our results indicate that bones, especially aged bones, provide an important source of a set of endocrine factors, including AdipoQ, that control body metabolism. © 2021 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Tejido Adiposo , Metabolismo Energético , Tejido Adiposo/metabolismo , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Osteoblastos/metabolismo
15.
Nat Commun ; 10(1): 5070, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31699980

RESUMEN

ß-Adrenergic receptor (ß-AR) signaling is a pathway controlling adaptive thermogenesis in brown or beige adipocytes. Here we investigate the biological roles of the transcription factor Foxp1 in brown/beige adipocyte differentiation and thermogenesis. Adipose-specific deletion of Foxp1 leads to an increase of brown adipose activity and browning program of white adipose tissues. The Foxp1-deficient mice show an augmented energy expenditure and are protected from diet-induced obesity and insulin resistance. Consistently, overexpression of Foxp1 in adipocytes impairs adaptive thermogenesis and promotes diet-induced obesity. A robust change in abundance of the ß3-adrenergic receptor (ß3-AR) is observed in brown/beige adipocytes from both lines of mice. Molecularly, Foxp1 directly represses ß3-AR transcription and regulates its desensitization behavior. Taken together, our findings reveal Foxp1 as a master transcriptional repressor of brown/beige adipocyte differentiation and thermogenesis, and provide an important clue for its targeting and treatment of obesity.


Asunto(s)
Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Adipogénesis/genética , Metabolismo Energético/genética , Factores de Transcripción Forkhead/genética , Receptores Adrenérgicos beta 3/genética , Proteínas Represoras/genética , Termogénesis/genética , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Prueba de Tolerancia a la Glucosa , Humanos , Resistencia a la Insulina , Ratones , Obesidad/genética , Obesidad/metabolismo , Epiplón/metabolismo , Feocromocitoma/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Proteínas Represoras/metabolismo
16.
J Clin Invest ; 127(4): 1241-1253, 2017 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-28240601

RESUMEN

A hallmark of aged mesenchymal stem/progenitor cells (MSCs) in bone marrow is the pivot of differentiation potency from osteoblast to adipocyte coupled with a decrease in self-renewal capacity. However, how these cellular events are orchestrated in the aging progress is not fully understood. In this study, we have used molecular and genetic approaches to investigate the role of forkhead box P1 (FOXP1) in transcriptional control of MSC senescence. In bone marrow MSCs, FOXP1 expression levels declined with age in an inverse manner with those of the senescence marker p16INK4A. Conditional depletion of Foxp1 in bone marrow MSCs led to premature aging characteristics, including increased bone marrow adiposity, decreased bone mass, and impaired MSC self-renewal capacity in mice. At the molecular level, FOXP1 regulated cell-fate choice of MSCs through interactions with the CEBPß/δ complex and recombination signal binding protein for immunoglobulin κ J region (RBPjκ), key modulators of adipogenesis and osteogenesis, respectively. Loss of p16INK4A in Foxp1-deficient MSCs partially rescued the defects in replication capacity and bone mass accrual. Promoter occupancy analyses revealed that FOXP1 directly represses transcription of p16INK4A. These results indicate that FOXP1 attenuates MSC senescence by orchestrating their cell-fate switch while maintaining their replicative capacity in a dose- and age-dependent manner.


Asunto(s)
Células de la Médula Ósea/metabolismo , Senescencia Celular/fisiología , Factores de Transcripción Forkhead/metabolismo , Células Madre Mesenquimatosas/metabolismo , Proteínas Represoras/metabolismo , Adipogénesis/fisiología , Animales , Células de la Médula Ósea/citología , Proteína beta Potenciadora de Unión a CCAAT/genética , Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Proteína delta de Unión al Potenciador CCAAT/genética , Proteína delta de Unión al Potenciador CCAAT/metabolismo , Línea Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Factores de Transcripción Forkhead/genética , Humanos , Células Madre Mesenquimatosas/citología , Ratones , Ratones Noqueados , Osteogénesis/fisiología , Proteínas Represoras/genética
17.
Front Plant Sci ; 7: 285, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27066014

RESUMEN

With increase of temperature, F o gradually rose in both WT and the mutant inactivated in the type 1 NAD(P)H dehydrogenase (NDH), a double mutant disrupted the genes of ndhJ and ndhK (ΔndhJK) or a triple mutant disrupted the genes of ndhC, ndhJ, and ndhK (ΔndhCJK). The temperature threshold of Fo rise was about 3-5°C lower in the mutants than in WT, indicating ΔndhJK and ΔndhCJK were more sensitive to elevated temperature. The F o rise after the threshold was slower and the reached maximal level was lower in the mutants than in WT, implying the chlororespiratory pathway was suppressed when NDH was inactivated. Meanwhile, the maximum quantum efficiency of photosystem II (PS II) (F v /F m) decreased to a similar extent below 50°C in WT and mutants. However, the decline was sharper in WT when temperature rose above 55°C, indicating a down regulation of PS II photochemical activity by the chlororespiratory pathway in response to elevated temperature. On the other hand, in the presence of n-propyl gallate, an inhibitor of plastid terminal oxidase (PTOX), the less evident increase in F o while the more decrease in F v /F m in ΔndhCJK than in WT after incubation at 50°C for 6 h suggest the increased sensitivity to heat stress when both NDH and chlororespiratory pathways are suppressed. Moreover, the net photosynthetic rate and photo-efficiency decreased more significantly in ΔndhJK than in WT under the heat stressed conditions. Compared to the light-oxidation of P700, the difference in the dark-reduction of P700(+) between WT and ndhJK disruptant was much less under the heat stressed conditions, implying significantly enhanced cyclic electron flow in light and the competition for electron from PQ between PTOX and photosystem I in the dark at the elevated temperature. Heat-stimulated expression of both NdhK and PTOX significantly increased in WT, while the expression of PTOX was less in ΔndhJK than in WT. Meanwhile, the amount of active form of Rubisco activase decreased much more in the mutant. The results suggest that chlororespiration and cyclic electron flow mediated by NDH may coordinate to alleviate the over-reduction of stroma, thus to keep operation of CO2 assimilation at certain extent under heat stress condition.

18.
Bone ; 84: 38-46, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26688275

RESUMEN

Wnt/ß-catenin signaling has been reported to contribute to the development of bone fibrous dysplasia. However, it remains unclear whether fibrocytes and immune cells are involved in this ß-catenin-mediated bone marrow fibrosis. In this study, we showed that constitutive activation of ß-catenin by Col1a1-Cre (3.6-kb) exhibited bone marrow fibrosis, featured with expanded populations of fibrocytes, myofibroblasts and osteoprogenitors. Lineage tracing and IHC examinations showed that Col3.6-Cre display Cre recombinase activity not only in osteoprogenitors, but also in monocyte-derived fibrocytes in the endosteal niches of bones. Additionally, ß-catenin stimulated the secretion of cytokines and pro-fibrotic signals in bone marrow, including GM-CSF, TGFß1 and VEGF. Consequently, the frequency of differentiated immature monocyte-derived dendritic cells and naïve T cells was markedly increased in the mutant bone marrow. These phenotypes were quite different from those following ß-catenin activation in mature osteoblasts driven by Col1a1-Cre (2.3-kb). Our findings suggested that a conserved pro-fibrotic signal cascade might underlie ß-catenin-mediated bone marrow fibrosis, involving TGFß1-enhanced fibrocyte activation and immunoregulatory responses. This study might shed new light on the understanding and development of a therapeutic strategy for bone fibrous dysplasia.


Asunto(s)
Médula Ósea/patología , Inmunomodulación , Osteocitos/patología , Células Madre/metabolismo , beta Catenina/metabolismo , Animales , Médula Ósea/metabolismo , Linaje de la Célula , Colágeno Tipo I/metabolismo , Fibrosis , Inflamación/patología , Integrasas/metabolismo , Ratones , Mutación/genética , Células Mieloides/metabolismo , Células Mieloides/patología , Miofibroblastos/patología , Osteocitos/metabolismo , Transducción de Señal , Linfocitos T/patología , Regulación hacia Arriba
19.
PLoS One ; 10(7): e0131674, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26171970

RESUMEN

Hair follicle stem cells (HFSCs) in the bugle circularly generate outer root sheath (ORS) through linear proliferation within limited cycles during anagen phases. However, the mechanisms controlling the pace of HFSC proliferation remain unclear. Here we revealed that Foxp1, a transcriptional factor, was dynamically relocated from the nucleus to the cytoplasm of HFSCs in phase transitions from anagen to catagen, coupled with the rise of oxidative stress. Mass spectrum analyses revealed that the S468 phosphorylation of Foxp1 protein was responsive to oxidative stress and affected its nucleocytoplasmic translocation. Foxp1 deficiency in hair follicles led to compromised ROS accrual and increased HFSC proliferation. And more, NAC treatment profoundly elongated the anagen duration and HFSC proliferation in Foxp1-deficient background. Molecularly, Foxp1 augmented ROS levels through suppression of Trx1-mediated reductive function, thereafter imposing the cell cycle arrest by modulating the activity of p19/p53 pathway. Our findings identify a novel role for Foxp1 in controlling HFSC proliferation with cellular dynamic location in response to oxidative stress during hair cycling.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Folículo Piloso/citología , Folículo Piloso/crecimiento & desarrollo , Estrés Oxidativo , Proteínas Represoras/metabolismo , Células Madre/citología , Transporte Activo de Núcleo Celular , Animales , Línea Celular , Núcleo Celular/metabolismo , Proliferación Celular , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Masculino , Ratones , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Fase S , Tiorredoxinas/metabolismo
20.
Bone ; 55(1): 258-67, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23334081

RESUMEN

Wnt signaling has important roles in embryonic bone development and postnatal bone remodeling, but inconsistent impact on bone property is observed in different genetic alterations of Lrp5 and ß-catenin. More importantly, it is still controversial whether Lrp5 regulate bone formation locally or globally through gut-derived serotonin. Here we explored the function of Wnt proteins in osteoblastic niche through inactivation of the Wntless (Wls) gene, which abrogates the secretion of Wnts. The depletion of Wls in osteoblast progenitor cells resulted in severe osteopenia with more profound defects in osteoblastogenesis, osteoclastogenesis and maintenance of bone marrow mesenchymal stem cells (BMSCs) compared to that observed in Lrp5 and ß-catenin mutants. These findings support the point of view that Wnt/Lrp5 signaling locally regulates bone mass accrual through multiple effects of osteoblastic Wnts on osteoblastic bone formation and osteoclastic bone resorption. Moreover, osteoblastic Wnts confer a niche role for maintenance of BMSCs, providing novel cues for the definition of BMSCs niche in bone marrow.


Asunto(s)
Células de la Médula Ósea/metabolismo , Remodelación Ósea , Células Madre Mesenquimatosas/metabolismo , Osteoblastos/metabolismo , Proteínas Wnt/metabolismo , Animales , Animales Recién Nacidos , Enfermedades Óseas Metabólicas/diagnóstico por imagen , Enfermedades Óseas Metabólicas/metabolismo , Enfermedades Óseas Metabólicas/patología , Células de la Médula Ósea/patología , Huesos/diagnóstico por imagen , Huesos/metabolismo , Huesos/patología , Diferenciación Celular , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Factor Estimulante de Colonias de Macrófagos/metabolismo , Células Madre Mesenquimatosas/patología , Ratones , Ratones Endogámicos C57BL , Tamaño de los Órganos , Osteoblastos/patología , Osteogénesis , Comunicación Paracrina , Ligando RANK/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Microtomografía por Rayos X
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA