Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.402
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 22(9): 1093-1106, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34282331

RESUMEN

Neutrophils display distinct gene expression patters depending on their developmental stage, activation state and tissue microenvironment. To determine the transcription factor networks that shape these responses in a mouse model, we integrated transcriptional and chromatin analyses of neutrophils during acute inflammation. We showed active chromatin remodeling at two transition stages: bone marrow-to-blood and blood-to-tissue. Analysis of differentially accessible regions revealed distinct sets of putative transcription factors associated with control of neutrophil inflammatory responses. Using ex vivo and in vivo approaches, we confirmed that RUNX1 and KLF6 modulate neutrophil maturation, whereas RELB, IRF5 and JUNB drive neutrophil effector responses and RFX2 and RELB promote survival. Interfering with neutrophil activation by targeting one of these factors, JUNB, reduced pathological inflammation in a mouse model of myocardial infarction. Therefore, our study represents a blueprint for transcriptional control of neutrophil responses in acute inflammation and opens possibilities for stage-specific therapeutic modulation of neutrophil function in disease.


Asunto(s)
Ensamble y Desensamble de Cromatina/genética , Inflamación/inmunología , Neutrófilos/inmunología , Activación Transcripcional/genética , Animales , Células CHO , Línea Celular , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Cricetulus , Femenino , Factores Reguladores del Interferón/metabolismo , Factor 6 Similar a Kruppel/metabolismo , Ratones , Ratones Endogámicos C57BL , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Factores de Transcripción del Factor Regulador X/metabolismo , Factor de Transcripción ReIB/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética/genética
2.
Cell ; 161(7): 1527-38, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26073941

RESUMEN

Dendritic cells (DCs) are required to initiate and sustain T cell-dependent anti-cancer immunity. However, tumors often evade immune control by crippling normal DC function. The endoplasmic reticulum (ER) stress response factor XBP1 promotes intrinsic tumor growth directly, but whether it also regulates the host anti-tumor immune response is not known. Here we show that constitutive activation of XBP1 in tumor-associated DCs (tDCs) drives ovarian cancer (OvCa) progression by blunting anti-tumor immunity. XBP1 activation, fueled by lipid peroxidation byproducts, induced a triglyceride biosynthetic program in tDCs leading to abnormal lipid accumulation and subsequent inhibition of tDC capacity to support anti-tumor T cells. Accordingly, DC-specific XBP1 deletion or selective nanoparticle-mediated XBP1 silencing in tDCs restored their immunostimulatory activity in situ and extended survival by evoking protective type 1 anti-tumor responses. Targeting the ER stress response should concomitantly inhibit tumor growth and enhance anti-cancer immunity, thus offering a unique approach to cancer immunotherapy.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Dendríticas/patología , Estrés del Retículo Endoplásmico , Neoplasias Ováricas/inmunología , Neoplasias Ováricas/patología , Factores de Transcripción/metabolismo , Animales , Femenino , Humanos , Peroxidación de Lípido , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Transcripción del Factor Regulador X , Linfocitos T/inmunología , Proteína 1 de Unión a la X-Box
3.
Cell ; 156(6): 1125-1127, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24630714

RESUMEN

The hexosamine biosynthetic pathway (HBP) generates metabolites for protein N- and O-glycosylation. Wang et al. and Denzel et al. report a hitherto unknown link between the HBP and stress in the endoplasmic reticulum. These studies establish the HBP as a critical component of the cellular machinery of protein homeostasis.


Asunto(s)
Vías Biosintéticas , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora)/metabolismo , Hexosaminas/metabolismo , Longevidad , Proteínas/metabolismo , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada , Animales , Humanos , Masculino , Factores de Transcripción del Factor Regulador X
4.
Cell ; 156(6): 1179-1192, 2014 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-24630721

RESUMEN

The hexosamine biosynthetic pathway (HBP) generates uridine diphosphate N-acetylglucosamine (UDP-GlcNAc) for glycan synthesis and O-linked GlcNAc (O-GlcNAc) protein modifications. Despite the established role of the HBP in metabolism and multiple diseases, regulation of the HBP remains largely undefined. Here, we show that spliced X-box binding protein 1 (Xbp1s), the most conserved signal transducer of the unfolded protein response (UPR), is a direct transcriptional activator of the HBP. We demonstrate that the UPR triggers HBP activation via Xbp1s-dependent transcription of genes coding for key, rate-limiting enzymes. We further establish that this previously unrecognized UPR-HBP axis is triggered in a variety of stress conditions. Finally, we demonstrate a physiologic role for the UPR-HBP axis by showing that acute stimulation of Xbp1s in heart by ischemia/reperfusion confers robust cardioprotection in part through induction of the HBP. Collectively, these studies reveal that Xbp1s couples the UPR to the HBP to protect cells under stress.


Asunto(s)
Vías Biosintéticas , Proteínas de Unión al ADN/metabolismo , Hexosaminas/metabolismo , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada , Animales , Glutamina-Fructosa-6-Fosfato Transaminasa (Isomerizadora) , Humanos , Masculino , Ratones , Ratones Transgénicos , Isquemia Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/metabolismo , Miocitos Cardíacos/metabolismo , Transferasas de Grupos Nitrogenados/genética , Factores de Transcripción del Factor Regulador X , Proteína 1 de Unión a la X-Box
5.
Nat Immunol ; 17(3): 323-30, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26779600

RESUMEN

Plasma cell differentiation requires silencing of B cell transcription, while it establishes antibody-secretory function and long-term survival. The transcription factors Blimp-1 and IRF4 are essential for the generation of plasma cells; however, their function in mature plasma cells has remained elusive. We found that while IRF4 was essential for the survival of plasma cells, Blimp-1 was dispensable for this. Blimp-1-deficient plasma cells retained their transcriptional identity but lost the ability to secrete antibody. Blimp-1 regulated many components of the unfolded protein response (UPR), including XBP-1 and ATF6. The overlap in the functions of Blimp-1 and XBP-1 was restricted to that response, with Blimp-1 uniquely regulating activity of the kinase mTOR and the size of plasma cells. Thus, Blimp-1 was required for the unique physiological ability of plasma cells that enables the secretion of protective antibody.


Asunto(s)
Diferenciación Celular/inmunología , Inmunoglobulinas/inmunología , Factores Reguladores del Interferón/inmunología , Células Plasmáticas/inmunología , Factores de Transcripción/inmunología , Respuesta de Proteína Desplegada/inmunología , Factor de Transcripción Activador 6/genética , Factor de Transcripción Activador 6/inmunología , Animales , Tamaño de la Célula , Inmunoprecipitación de Cromatina , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunoglobulinas/metabolismo , Factores Reguladores del Interferón/genética , Ratones , Ratones Noqueados , Microscopía Electrónica de Transmisión , Células Plasmáticas/metabolismo , Factor 1 de Unión al Dominio 1 de Regulación Positiva , Factores de Transcripción del Factor Regulador X , Análisis de Secuencia de ADN , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Factores de Transcripción/genética , Respuesta de Proteína Desplegada/genética , Proteína 1 de Unión a la X-Box
6.
Cell ; 152(5): 1146-59, 2013 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-23434322

RESUMEN

Tet proteins oxidize 5-methylcytosine (mC) to generate 5-hydroxymethyl (hmC), 5-formyl (fC), and 5-carboxylcytosine (caC). The exact function of these oxidative cytosine bases remains elusive. We applied quantitative mass-spectrometry-based proteomics to identify readers for mC and hmC in mouse embryonic stem cells (mESC), neuronal progenitor cells (NPC), and adult mouse brain tissue. Readers for these modifications are only partially overlapping, and some readers, such as Rfx proteins, display strong specificity. Interactions are dynamic during differentiation, as for example evidenced by the mESC-specific binding of Klf4 to mC and the NPC-specific binding of Uhrf2 to hmC, suggesting specific biological roles for mC and hmC. Oxidized derivatives of mC recruit distinct transcription regulators as well as a large number of DNA repair proteins in mouse ES cells, implicating the DNA damage response as a major player in active DNA demethylation.


Asunto(s)
5-Metilcitosina/análisis , Citosina/análogos & derivados , Metilación de ADN , 5-Metilcitosina/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Citosina/análisis , Citosina/metabolismo , ADN Glicosilasas/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Madre Embrionarias/metabolismo , Factor 4 Similar a Kruppel , Espectrometría de Masas , Ratones , Oxidación-Reducción , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción del Factor Regulador X , Células Madre/metabolismo , Factores de Transcripción/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
7.
Development ; 151(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38722098

RESUMEN

During development, the gastrointestinal tract undergoes patterning along its anterior-posterior axis to define regions with distinct organs and functions. A new paper in Development derives human intestinal organoids from an individual with duodenal defects and a compound heterozygous variant in the gene encoding the transcription factor RFX6. By studying these organoids, the authors identify novel roles for RFX6 in intestinal patterning. To learn more about the story behind the paper, we caught up with first author J. Guillermo Sanchez and corresponding author Jim Wells, an endowed professor in the Division of Developmental Biology at Cincinnati Children's Hospital, USA, where he is also the Director for Basic Research in the Division of Endocrinology.


Asunto(s)
Biología Evolutiva , Humanos , Historia del Siglo XXI , Historia del Siglo XX , Biología Evolutiva/historia , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Organoides/metabolismo , Tipificación del Cuerpo/genética
8.
Development ; 151(9)2024 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-38587174

RESUMEN

The gastrointestinal (GI) tract is complex and consists of multiple organs with unique functions. Rare gene variants can cause congenital malformations of the human GI tract, although the molecular basis of these has been poorly studied. We identified a patient with compound-heterozygous variants in RFX6 presenting with duodenal malrotation and atresia, implicating RFX6 in development of the proximal intestine. To identify how mutations in RFX6 impact intestinal patterning and function, we derived induced pluripotent stem cells from this patient to generate human intestinal organoids (HIOs). We identified that the duodenal HIOs and human tissues had mixed regional identity, with gastric and ileal features. CRISPR-mediated correction of RFX6 restored duodenal identity. We then used gain- and loss-of-function and transcriptomic approaches in HIOs and Xenopus embryos to identify that PDX1 is a downstream transcriptional target of RFX6 required for duodenal development. However, RFX6 had additional PDX1-independent transcriptional targets involving multiple components of signaling pathways that are required for establishing early regional identity in the GI tract. In summary, we have identified RFX6 as a key regulator in intestinal patterning that acts by regulating transcriptional and signaling pathways.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas de Homeodominio , Organoides , Factores de Transcripción del Factor Regulador X , Transactivadores , Humanos , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Animales , Proteínas de Homeodominio/metabolismo , Proteínas de Homeodominio/genética , Transactivadores/metabolismo , Transactivadores/genética , Organoides/metabolismo , Organoides/embriología , Duodeno/metabolismo , Duodeno/embriología , Intestinos/embriología , Atresia Intestinal/genética , Células Madre Pluripotentes Inducidas/metabolismo , Tipificación del Cuerpo/genética , Transducción de Señal/genética , Mutación/genética
9.
Nat Immunol ; 16(8): 829-37, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26147683

RESUMEN

The transcription factor XBP1 has been linked to the development of highly secretory tissues such as plasma cells and Paneth cells, yet its function in granulocyte maturation has remained unknown. Here we discovered an unexpectedly selective and absolute requirement for XBP1 in eosinophil differentiation without an effect on the survival of basophils or neutrophils. Progenitors of myeloid cells and eosinophils selectively activated the endoribonuclease IRE1α and spliced Xbp1 mRNA without inducing parallel endoplasmic reticulum (ER) stress signaling pathways. Without XBP1, nascent eosinophils exhibited massive defects in the post-translational maturation of key granule proteins required for survival, and these unresolvable structural defects fed back to suppress critical aspects of the transcriptional developmental program. Hence, we present evidence that granulocyte subsets can be distinguished by their differential reliance on secretory-pathway homeostasis.


Asunto(s)
Diferenciación Celular/inmunología , Proteínas de Unión al ADN/inmunología , Eosinófilos/inmunología , Expresión Génica/inmunología , Factores de Transcripción/inmunología , Animales , Diferenciación Celular/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico/genética , Estrés del Retículo Endoplásmico/inmunología , Endorribonucleasas/genética , Endorribonucleasas/inmunología , Endorribonucleasas/metabolismo , Eosinófilos/metabolismo , Eosinófilos/ultraestructura , Citometría de Flujo , Perfilación de la Expresión Génica , Células Precursoras de Granulocitos/inmunología , Células Precursoras de Granulocitos/metabolismo , Células Precursoras de Granulocitos/ultraestructura , Células HEK293 , Humanos , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Microscopía Confocal , Microscopía Electrónica de Transmisión , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción del Factor Regulador X , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/inmunología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
10.
Nat Immunol ; 15(3): 248-57, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24441789

RESUMEN

The role of the unfolded protein response (UPR) and endoplasmic reticulum (ER) stress in homeostasis of the immune system is incompletely understood. Here we found that dendritic cells (DCs) constitutively activated the UPR sensor IRE-1α and its target, the transcription factor XBP-1, in the absence of ER stress. Loss of XBP-1 in CD11c+ cells led to defects in phenotype, ER homeostasis and antigen presentation by CD8α+ conventional DCs, yet the closely related CD11b+ DCs were unaffected. Whereas the dysregulated ER in XBP-1-deficient DCs resulted from loss of XBP-1 transcriptional activity, the phenotypic and functional defects resulted from regulated IRE-1α-dependent degradation (RIDD) of mRNAs, including those encoding CD18 integrins and components of the major histocompatibility complex (MHC) class I machinery. Thus, a precisely regulated feedback circuit involving IRE-1α and XBP-1 controls the homeostasis of CD8α+ conventional DCs.


Asunto(s)
Reactividad Cruzada/inmunología , Proteínas de Unión al ADN/inmunología , Células Dendríticas/inmunología , Endorribonucleasas/inmunología , Proteínas Serina-Treonina Quinasas/inmunología , Desplegamiento Proteico , Factores de Transcripción/inmunología , Respuesta de Proteína Desplegada/inmunología , Animales , Presentación de Antígeno/inmunología , Antígenos CD8/inmunología , Antígenos CD8/metabolismo , Proteínas de Unión al ADN/metabolismo , Células Dendríticas/metabolismo , Retículo Endoplásmico/inmunología , Endorribonucleasas/metabolismo , Retroalimentación Fisiológica/fisiología , Homeostasis/inmunología , Immunoblotting , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción del Factor Regulador X , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/metabolismo , Proteína 1 de Unión a la X-Box
11.
PLoS Biol ; 20(2): e3001538, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-35143476

RESUMEN

Within the immune system, microRNAs (miRNAs) exert key regulatory functions. However, what are the mRNA targets regulated by miRNAs and how miRNAs are transcriptionally regulated themselves remain for the most part unknown. We found that in primary human memory T helper lymphocytes, miR-150 was the most abundantly expressed miRNA, and its expression decreased drastically upon activation, suggesting regulatory roles. Constitutive MIR150 gene expression required the RFX family of transcription factors, and its activation-induced down-regulation was linked to their reduced expression. By performing miRNA pull-down and sequencing experiments, we identified PDGFA-associated protein 1 (PDAP1) as one main target of miR-150 in human T lymphocytes. PDAP1 acted as an RNA-binding protein (RBP), and its CRISPR/Cas-9-mediated deletion revealed that it prominently contributed to the regulation of T-cell proliferation. Overall, using an integrated approach involving quantitative analysis, unbiased genomics, and genome editing, we identified RFX factors, miR-150, and the PDAP1 RBP as the components of a regulatory axis that restrains proliferation of primary human T lymphocytes.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Proliferación Celular/genética , Regulación de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/genética , MicroARNs/genética , Factores de Transcripción del Factor Regulador X/genética , Regiones no Traducidas 3'/genética , Western Blotting , Linfocitos T CD4-Positivos/citología , Células Cultivadas , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Células HEK293 , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Células Jurkat , Activación de Linfocitos/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
12.
Cell Mol Life Sci ; 81(1): 99, 2024 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-38386071

RESUMEN

Proneural genes play a crucial role in neuronal differentiation. However, our understanding of the regulatory mechanisms governing proneural genes during neuronal differentiation remains limited. RFX4, identified as a candidate regulator of proneural genes, has been reported to be associated with the development of neuropsychiatric disorders. To uncover the regulatory relationship, we utilized a combination of multi-omics data, including ATAC-seq, ChIP-seq, Hi-C, and RNA-seq, to identify RFX4 as an upstream regulator of proneural genes. We further validated the role of RFX4 using an in vitro model of neuronal differentiation with RFX4 knock-in and a CRISPR-Cas9 knock-out system. As a result, we found that RFX4 directly interacts with the promoters of POU3F2 and NEUROD1. Transcriptomic analysis revealed a set of genes associated with neuronal development, which are highly implicated in the development of neuropsychiatric disorders, including schizophrenia. Notably, ectopic expression of RFX4 can drive human embryonic stem cells toward a neuronal fate. Our results strongly indicate that RFX4 serves as a direct upstream regulator of proneural genes, a role that is essential for normal neuronal development. Impairments in RFX4 function could potentially be related to the development of various neuropsychiatric disorders. However, understanding the precise mechanisms by which the RFX4 gene influences the onset of neuropsychiatric disorders requires further investigation through human genetic studies.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas de Homeodominio , Neuronas , Factores del Dominio POU , Factores de Transcripción del Factor Regulador X , Humanos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Perfilación de la Expresión Génica , Regiones Promotoras Genéticas , RNA-Seq , Diferenciación Celular , Proteínas de Homeodominio/genética , Factores del Dominio POU/genética , Factores de Transcripción del Factor Regulador X/genética
13.
Diabetologia ; 67(8): 1642-1662, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38743124

RESUMEN

AIMS/HYPOTHESIS: Regulatory factor X 6 (RFX6) is crucial for pancreatic endocrine development and differentiation. The RFX6 variant p.His293LeufsTer7 is significantly enriched in the Finnish population, with almost 1:250 individuals as a carrier. Importantly, the FinnGen study indicates a high predisposition for heterozygous carriers to develop type 2 and gestational diabetes. However, the precise mechanism of this predisposition remains unknown. METHODS: To understand the role of this variant in beta cell development and function, we used CRISPR technology to generate allelic series of pluripotent stem cells. We created two isogenic stem cell models: a human embryonic stem cell model; and a patient-derived stem cell model. Both were differentiated into pancreatic islet lineages (stem-cell-derived islets, SC-islets), followed by implantation in immunocompromised NOD-SCID-Gamma mice. RESULTS: Stem cell models of the homozygous variant RFX6-/- predictably failed to generate insulin-secreting pancreatic beta cells, mirroring the phenotype observed in Mitchell-Riley syndrome. Notably, at the pancreatic endocrine stage, there was an upregulation of precursor markers NEUROG3 and SOX9, accompanied by increased apoptosis. Intriguingly, heterozygous RFX6+/- SC-islets exhibited RFX6 haploinsufficiency (54.2% reduction in protein expression), associated with reduced beta cell maturation markers, altered calcium signalling and impaired insulin secretion (62% and 54% reduction in basal and high glucose conditions, respectively). However, RFX6 haploinsufficiency did not have an impact on beta cell number or insulin content. The reduced insulin secretion persisted after in vivo implantation in mice, aligning with the increased risk of variant carriers to develop diabetes. CONCLUSIONS/INTERPRETATION: Our allelic series isogenic SC-islet models represent a powerful tool to elucidate specific aetiologies of diabetes in humans, enabling the sensitive detection of aberrations in both beta cell development and function. We highlight the critical role of RFX6 in augmenting and maintaining the pancreatic progenitor pool, with an endocrine roadblock and increased cell death upon its loss. We demonstrate that RFX6 haploinsufficiency does not affect beta cell number or insulin content but does impair function, predisposing heterozygous carriers of loss-of-function variants to diabetes. DATA AVAILABILITY: Ultra-deep bulk RNA-seq data for pancreatic differentiation stages 3, 5 and 7 of H1 RFX6 genotypes are deposited in the Gene Expression Omnibus database with accession code GSE234289. Original western blot images are deposited at Mendeley ( https://data.mendeley.com/datasets/g75drr3mgw/2 ).


Asunto(s)
Haploinsuficiencia , Células Secretoras de Insulina , Factores de Transcripción del Factor Regulador X , Células Secretoras de Insulina/metabolismo , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/metabolismo , Animales , Humanos , Ratones , Diferenciación Celular/genética , Ratones Endogámicos NOD , Ratones SCID , Predisposición Genética a la Enfermedad , Femenino , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Proteínas del Tejido Nervioso
14.
Nat Immunol ; 13(10): 972-80, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22961053

RESUMEN

The transcription factor Foxp3 participates dominantly in the specification and function of Foxp3(+)CD4(+) regulatory T cells (T(reg) cells) but is neither strictly necessary nor sufficient to determine the characteristic T(reg) cell signature. Here we used computational network inference and experimental testing to assess the contribution of other transcription factors to this. Enforced expression of Helios or Xbp1 elicited distinct signatures, but Eos, IRF4, Satb1, Lef1 and GATA-1 elicited exactly the same outcome, acting in synergy with Foxp3 to activate expression of most of the T(reg) cell signature, including key transcription factors, and enhancing occupancy by Foxp3 at its genomic targets. Conversely, the T(reg) cell signature was robust after inactivation of any single cofactor. A redundant genetic switch thus 'locked in' the T(reg) cell phenotype, a model that would account for several aspects of T(reg) cell physiology, differentiation and stability.


Asunto(s)
Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Antígenos CD4/biosíntesis , Diferenciación Celular , Biología Computacional , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Factor de Transcripción GATA1/genética , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/genética , Factores Reguladores del Interferón/genética , Activación de Linfocitos , Factor de Unión 1 al Potenciador Linfoide/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción del Factor Regulador X , Serina Endopeptidasas/genética , Linfocitos T Reguladores/citología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteína 1 de Unión a la X-Box
15.
Am J Med Genet A ; 194(12): e63816, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-39007708

RESUMEN

RFX7 encodes a transcription factor that is ubiquitously expressed and important for neural development. Haploinsufficiency of RFX7 is associated with intellectual disability, developmental delay, and diverse malformations of brain structures. Currently, there are only 16 clinically described individuals who have variants in RFX7. A recognizable pattern of malformation associated with mutation in RFX7 has not yet been uncovered. Here we describe the phenotypic presentation of two additional individuals who have novel de novo variants in RFX7. One of the individuals we describe is from an under-represented Afro-Caribbean population.


Asunto(s)
Mutación , Fenotipo , Humanos , Masculino , Femenino , Mutación/genética , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Factores de Transcripción del Factor Regulador X/genética , Niño , Discapacidades del Desarrollo/genética , Discapacidades del Desarrollo/patología , Preescolar , Haploinsuficiencia/genética , Lactante , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad
16.
Immunity ; 43(3): 451-62, 2015 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-26341399

RESUMEN

Endoplasmic reticulum (ER) stress is observed in many human diseases, often associated with inflammation. ER stress can trigger inflammation through nucleotide-binding domain and leucine-rich repeat containing (NLRP3) inflammasome, which might stimulate inflammasome formation by association with damaged mitochondria. How ER stress triggers mitochondrial dysfunction and inflammasome activation is ill defined. Here we have used an infection model to show that the IRE1α ER stress sensor regulates regulated mitochondrial dysfunction through an NLRP3-mediated feed-forward loop, independently of ASC. IRE1α activation increased mitochondrial reactive oxygen species, promoting NLRP3 association with mitochondria. NLRP3 was required for ER stress-induced cleavage of caspase-2 and the pro-apoptotic factor, Bid, leading to subsequent release of mitochondrial contents. Caspase-2 and Bid were necessary for activation of the canonical inflammasome by infection-associated or general ER stress. These data identify an NLRP3-caspase-2-dependent mechanism that relays ER stress to the mitochondria to promote inflammation, integrating cellular stress and innate immunity.


Asunto(s)
Proteínas Portadoras/inmunología , Caspasa 2/inmunología , Estrés del Retículo Endoplásmico/inmunología , Inflamasomas/inmunología , Mitocondrias/inmunología , Animales , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/genética , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/inmunología , Proteína Proapoptótica que Interacciona Mediante Dominios BH3/metabolismo , Western Blotting , Brucella abortus/inmunología , Brucella abortus/fisiología , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Caspasa 2/genética , Caspasa 2/metabolismo , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Estrés del Retículo Endoplásmico/genética , Endorribonucleasas/inmunología , Endorribonucleasas/metabolismo , Células HEK293 , Interacciones Huésped-Patógeno/inmunología , Humanos , Inflamasomas/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/genética , Mitocondrias/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR , Proteínas Serina-Treonina Quinasas/inmunología , Proteínas Serina-Treonina Quinasas/metabolismo , Interferencia de ARN/inmunología , Especies Reactivas de Oxígeno/inmunología , Especies Reactivas de Oxígeno/metabolismo , Factores de Transcripción del Factor Regulador X , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
17.
Proc Natl Acad Sci U S A ; 118(9)2021 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-33622787

RESUMEN

HLA-C arose during evolution of pregnancy in the great apes 10 to 15 million years ago. It has a dual function on placental extravillous trophoblasts (EVTs) as it contributes to both tolerance and immunity at the maternal-fetal interface. The mode of its regulation is of considerable interest in connection with the biology of pregnancy and pregnancy abnormalities. First-trimester primary EVTs in which HLA-C is highly expressed, as well as JEG3, an EVT model cell line, were employed. Single-cell RNA-seq data and quantitative PCR identified high expression of the transcription factor ELF3 in those cells. Chromatin immunoprecipitation (ChIP)-PCR confirmed that both ELF3 and MED1 bound to the proximal HLA-C promoter region. However, binding of RFX5 to this region was absent or severely reduced, and the adjacent HLA-B locus remained closed. Expression of HLA-C was inhibited by ELF3 small interfering RNAs (siRNAs) and by wrenchnolol treatment. Wrenchnolol is a cell-permeable synthetic organic molecule that mimics ELF3 and is relatively specific for binding to ELF3's coactivator, MED23, as our data also showed in JEG3. Moreover, the ELF3 gene is regulated by a superenhancer that spans more than 5 Mb, identified by assay for transposase-accessible chromatin using sequencing (ATAC-seq), as well as by its sensitivity to (+)-JQ1 (inhibitor of BRD4). ELF3 bound to its own promoter, thus creating an autoregulatory feedback loop that establishes expression of ELF3 and HLA-C in trophoblasts. Wrenchnolol blocked binding of MED23 to ELF3, thus disrupting the positive-feedback loop that drives ELF3 expression, with down-regulation of HLA-C expression as a consequence.


Asunto(s)
Proteínas de Unión al ADN/genética , Elementos de Facilitación Genéticos , Retroalimentación Fisiológica , Antígenos HLA-C/genética , Proteínas Proto-Oncogénicas c-ets/genética , Factores de Transcripción/genética , Trofoblastos/inmunología , Aborto Legal , Adamantano/farmacología , Azepinas/farmacología , Línea Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/inmunología , Femenino , Regulación del Desarrollo de la Expresión Génica/inmunología , Antígenos HLA-B/genética , Antígenos HLA-B/inmunología , Antígenos HLA-C/inmunología , Humanos , Inmunidad Materno-Adquirida , Indoles/farmacología , Complejo Mediador/genética , Complejo Mediador/inmunología , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/inmunología , Embarazo , Primer Trimestre del Embarazo , Cultivo Primario de Células , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Proto-Oncogénicas c-ets/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-ets/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Factores de Transcripción del Factor Regulador X/genética , Factores de Transcripción del Factor Regulador X/inmunología , Transducción de Señal , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/inmunología , Triazoles/farmacología , Trofoblastos/citología , Trofoblastos/efectos de los fármacos
18.
Gastroenterology ; 162(3): 799-812, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34687740

RESUMEN

BACKGROUND & AIMS: A detailed understanding of antitumor immunity is essential for optimal cancer immune therapy. Although defective mutations in the B2M and HLA-ABC genes, which encode molecules essential for antigen presentation, have been reported in several studies, the effects of these defects on tumor immunity have not been quantitatively evaluated. METHODS: Mutations in HLA-ABC genes were analyzed in 114 microsatellite instability-high colorectal cancers using a long-read sequencer. The data were further analyzed in combination with whole-exome sequencing, transcriptome sequencing, DNA methylation array, and immunohistochemistry data. RESULTS: We detected 101 truncating mutations in 57 tumors (50%) and loss of 61 alleles in 21 tumors (18%). Based on the integrated analysis that enabled the immunologic subclassification of microsatellite instability-high colorectal cancers, we identified a subtype of tumors in which lymphocyte infiltration was reduced, partly due to reduced expression of HLA-ABC genes in the absence of apparent genetic alterations. Survival time of patients with such tumors was shorter than in patients with other tumor types. Paradoxically, tumor mutation burden was highest in the subtype, suggesting that the immunogenic effect of accumulating mutations was counterbalanced by mutations that weakened immunoreactivity. Various genetic and epigenetic alterations, including frameshift mutations in RFX5 and promoter methylation of PSMB8 and HLA-A, converged on reduced expression of HLA-ABC genes. CONCLUSIONS: Our detailed immunogenomic analysis provides information that will facilitate the improvement and development of cancer immunotherapy.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/inmunología , Genes MHC Clase I/genética , Escape del Tumor/genética , Escape del Tumor/inmunología , Microglobulina beta-2/genética , Alelos , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Metilación de ADN , Epigénesis Genética , Expresión Génica , Antígenos HLA-A/genética , Antígenos HLA-A/metabolismo , Humanos , Inmunogenética , Linfocitos Infiltrantes de Tumor , Inestabilidad de Microsatélites , Complejo de la Endopetidasa Proteasomal/genética , Factores de Transcripción del Factor Regulador X/genética , Tasa de Supervivencia , Microglobulina beta-2/metabolismo
19.
Development ; 147(21)2020 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-33033118

RESUMEN

Mitchell-Riley syndrome (MRS) is caused by recessive mutations in the regulatory factor X6 gene (RFX6) and is characterised by pancreatic hypoplasia and neonatal diabetes. To determine why individuals with MRS specifically lack pancreatic endocrine cells, we micro-CT imaged a 12-week-old foetus homozygous for the nonsense mutation RFX6 c.1129C>T, which revealed loss of the pancreas body and tail. From this foetus, we derived iPSCs and show that differentiation of these cells in vitro proceeds normally until generation of pancreatic endoderm, which is significantly reduced. We additionally generated an RFX6HA reporter allele by gene targeting in wild-type H9 cells to precisely define RFX6 expression and in parallel performed in situ hybridisation for RFX6 in the dorsal pancreatic bud of a Carnegie stage 14 human embryo. Both in vitro and in vivo, we find that RFX6 specifically labels a subset of PDX1-expressing pancreatic endoderm. In summary, RFX6 is essential for efficient differentiation of pancreatic endoderm, and its absence in individuals with MRS specifically impairs formation of endocrine cells of the pancreas head and tail.


Asunto(s)
Diferenciación Celular , Diabetes Mellitus/genética , Diabetes Mellitus/patología , Endodermo/embriología , Enfermedades de la Vesícula Biliar/genética , Enfermedades de la Vesícula Biliar/patología , Células Madre Pluripotentes Inducidas/patología , Atresia Intestinal/genética , Atresia Intestinal/patología , Mutación/genética , Páncreas/embriología , Factores de Transcripción del Factor Regulador X/genética , Alelos , Secuencia de Bases , Diferenciación Celular/genética , Cromatina/metabolismo , Consanguinidad , Diabetes Mellitus/diagnóstico por imagen , Embrión de Mamíferos/metabolismo , Desarrollo Embrionario , Familia , Femenino , Enfermedades de la Vesícula Biliar/diagnóstico por imagen , Genoma Humano , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Atresia Intestinal/diagnóstico por imagen , Masculino , Linaje , Transcripción Genética , Transcriptoma/genética , Microtomografía por Rayos X
20.
J Neurooncol ; 165(3): 487-497, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38057505

RESUMEN

PURPOSE: Spinal ependymoma (SE) is a rare tumor that is most commonly low-grade and tends to recur when complete tumor resection is not feasible. We investigated the molecular mechanism induces stem cell features in SE. METHODS: Immunohistochemical staining was conducted to analyze the expression of RFX2 in tumor tissues of SE patients at different stages. The expression of tumor stemness markers (Netsin and CD133) was analyzed using western blot analysis and IF, and the efficiency of sphere formation in SE cells was analyzed. The biological activities of SE cells were analyzed by EdU proliferation assay, TUNEL, wound healing, and Transwell assays. The regulatory relationship of RFX2 on PAF1 was verified by ChIP-qPCR and the dual-luciferase assay. SE cells were injected into the spinal cord of nude mice for in vivo assays. RESULTS: RFX2 was higher in the tumor tissues of SE-III patients than in the tumor tissues of SE-I patients. RFX2 knockdown reduced the expression of tumor stemness markers in SE cells and inhibited the sphere formation efficiency. Moreover, RFX2 knockdown ameliorated the malignant progression of SE in nude mice, as manifested by prolonged survival and alleviated SE tumor infiltration. RFX2 bound to the PAF1 promoter to induce its transcription. Overexpression of PAF1 overturned the effects of RFX2 knockdown on stem cell features and biological activities of SE cells, thereby reducing survival in mice. CONCLUSIONS: RFX2 activates PAF1 transcription, which promotes tumor stemness of SE cells and leads to the malignant progression of SE.


Asunto(s)
Ependimoma , Epigénesis Genética , Humanos , Animales , Ratones , Ratones Desnudos , Línea Celular Tumoral , Recurrencia Local de Neoplasia/patología , Ependimoma/patología , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Células Madre Neoplásicas/patología , Factores de Transcripción del Factor Regulador X/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA