Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.055
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 19(7): 665-673, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29925983

RESUMEN

In recent years, the understanding of regulatory T cell (Treg cell) biology has expanded considerably. Key observations have challenged the traditional definition of Treg cells and have provided insight into the underlying mechanisms responsible for the development of autoimmune diseases, with new therapeutic strategies that improve disease outcome. This Review summarizes the newer concepts of Treg cell instability, Treg cell plasticity and tissue-specific Treg cells, and their relationship to autoimmunity. Those three main concepts have changed the understanding of Treg cell biology: how they interact with other immune and non-immune cells; their functions in specific tissues; and the implications of this for the pathogenesis of autoimmune diseases.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Linfocitos T Reguladores/inmunología , Animales , Factores de Transcripción Forkhead/fisiología , Humanos , Ratones
2.
Genes Dev ; 34(15-16): 1039-1050, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32561546

RESUMEN

The FoxA transcription factors are critical for liver development through their pioneering activity, which initiates a highly complex regulatory network thought to become progressively resistant to the loss of any individual hepatic transcription factor via mutual redundancy. To investigate the dispensability of FoxA factors for maintaining this regulatory network, we ablated all FoxA genes in the adult mouse liver. Remarkably, loss of FoxA caused rapid and massive reduction in the expression of critical liver genes. Activity of these genes was reduced back to the low levels of the fetal prehepatic endoderm stage, leading to necrosis and lethality within days. Mechanistically, we found FoxA proteins to be required for maintaining enhancer activity, chromatin accessibility, nucleosome positioning, and binding of HNF4α. Thus, the FoxA factors act continuously, guarding hepatic enhancer activity throughout adult life.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Redes Reguladoras de Genes , Hígado/metabolismo , Animales , Sitios de Unión , Cromatina/metabolismo , Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica , Técnicas de Silenciamiento del Gen , Factor Nuclear 3-alfa del Hepatocito/genética , Factor Nuclear 3-beta del Hepatocito/genética , Factor Nuclear 3-gamma del Hepatocito/genética , Factor Nuclear 4 del Hepatocito/metabolismo , Hígado/patología , Fallo Hepático/etiología , Fallo Hepático/patología , Masculino , Ratones , Nucleosomas
3.
Nat Immunol ; 15(7): 667-75, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24859450

RESUMEN

CD4(+) follicular helper T cells (T(FH) cells) are essential for germinal center (GC) responses and long-lived antibody responses. Here we report that naive CD4(+) T cells deficient in the transcription factor Foxp1 'preferentially' differentiated into T(FH) cells, which resulted in substantially enhanced GC and antibody responses. We found that Foxp1 used both constitutive Foxp1A and Foxp1D induced by stimulation of the T cell antigen receptor (TCR) to inhibit the generation of T(FH) cells. Mechanistically, Foxp1 directly and negatively regulated interleukin 21 (IL-21); Foxp1 also dampened expression of the costimulatory molecule ICOS and its downstream signaling at early stages of T cell activation, which rendered Foxp1-deficient CD4(+) T cells partially resistant to blockade of the ICOS ligand (ICOSL) during T(FH) cell development. Our findings demonstrate that Foxp1 is a critical negative regulator of T(FH) cell differentiation.


Asunto(s)
Diferenciación Celular , Factores de Transcripción Forkhead/fisiología , Proteínas Represoras/fisiología , Linfocitos T Colaboradores-Inductores/citología , Animales , Linfocitos T CD4-Positivos/inmunología , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Interleucinas/genética , Ratones , Ratones Endogámicos C57BL , Receptores de Antígenos de Linfocitos T/fisiología
4.
Nat Immunol ; 15(7): 657-66, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24859451

RESUMEN

Follicular helper T cells (T(FH) cells) are responsible for effective B cell-mediated immunity, and Bcl-6 is a central factor for the differentiation of T(FH) cells. However, the molecular mechanisms that regulate the induction of T(FH) cells remain unclear. Here we found that the E3 ubiquitin ligase Itch was essential for the differentiation of T(FH) cells, germinal center responses and immunoglobulin G (IgG) responses to acute viral infection. Itch acted intrinsically in CD4(+) T cells at early stages of T(FH) cell development. Itch seemed to act upstream of Bcl-6 expression, as Bcl-6 expression was substantially impaired in Itch(-/-) cells, and the differentiation of Itch(-/-) T cells into T(FH) cells was restored by enforced expression of Bcl-6. Itch associated with the transcription factor Foxo1 and promoted its ubiquitination and degradation. The defective T(FH) differentiation of Itch(-/-) T cells was rectified by deletion of Foxo1. Thus, our results indicate that Itch acts as an essential positive regulator in the differentiation of T(FH) cells.


Asunto(s)
Diferenciación Celular , Linfocitos T Colaboradores-Inductores/citología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/fisiología , Centro Germinal/inmunología , Interleucina-2/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-bcl-6/fisiología , Células Th2/inmunología
5.
Nat Rev Mol Cell Biol ; 14(2): 83-97, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23325358

RESUMEN

Forkhead box O (FOXO) transcription factors are involved in the regulation of the cell cycle, apoptosis and metabolism. In model organisms, FOXO activity also affects stem cell maintenance and lifespan as well as age-related diseases, such as cancer and diabetes. Multiple upstream pathways regulate FOXO activity through post-translational modifications and nuclear-cytoplasmic shuttling of both FOXO and its regulators. The diversity of this upstream regulation and the downstream effects of FOXOs suggest that they function as homeostasis regulators to maintain tissue homeostasis over time and coordinate a response to environmental changes, including growth factor deprivation, metabolic stress (starvation) and oxidative stress.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Homeostasis/genética , Factores de Transcripción Forkhead/química , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Homeostasis/fisiología , Humanos , Modelos Biológicos , Estrés Oxidativo/genética , Estrés Oxidativo/fisiología , Procesamiento Proteico-Postraduccional/fisiología , Transducción de Señal/genética , Estrés Fisiológico/genética , Estrés Fisiológico/fisiología , Relación Estructura-Actividad
6.
Genes Dev ; 31(20): 2039-2055, 2017 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-29138280

RESUMEN

The molecular mechanisms driving brain development at risk in autism spectrum disorders (ASDs) remain mostly unknown. Previous studies have implicated the transcription factor FOXP1 in both brain development and ASD pathophysiology. However, the specific molecular pathways both upstream of and downstream from FOXP1 are not fully understood. To elucidate the contribution of FOXP1-mediated signaling to brain development and, in particular, neocortical development, we generated forebrain-specific Foxp1 conditional knockout mice. We show that deletion of Foxp1 in the developing forebrain leads to impairments in neonatal vocalizations as well as neocortical cytoarchitectonic alterations via neuronal positioning and migration. Using a genomics approach, we identified the transcriptional networks regulated by Foxp1 in the developing neocortex and found that such networks are enriched for downstream targets involved in neurogenesis and neuronal migration. We also uncovered mechanistic insight into Foxp1 function by demonstrating that sumoylation of Foxp1 during embryonic brain development is necessary for mediating proper interactions between Foxp1 and the NuRD complex. Furthermore, we demonstrated that sumoylation of Foxp1 affects neuronal differentiation and migration in the developing neocortex. Together, these data provide critical mechanistic insights into the function of FOXP1 in the developing neocortex and may reveal molecular pathways at risk in ASD.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Prosencéfalo/crecimiento & desarrollo , Proteínas Represoras/fisiología , Vocalización Animal , Animales , Movimiento Celular , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Eliminación de Gen , Expresión Génica , Complejo Desacetilasa y Remodelación del Nucleosoma Mi-2/metabolismo , Ratones Noqueados , Neocórtex/citología , Neocórtex/crecimiento & desarrollo , Neocórtex/metabolismo , Neuritas/fisiología , Neuronas/fisiología , Prosencéfalo/citología , Prosencéfalo/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Transducción de Señal , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Sumoilación
7.
PLoS Biol ; 19(4): e3001204, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33891586

RESUMEN

Many cell types display the remarkable ability to alter their cellular phenotype in response to specific external or internal signals. Such phenotypic plasticity is apparent in the nematode Caenorhabditis elegans when adverse environmental conditions trigger entry into the dauer diapause stage. This entry is accompanied by structural, molecular, and functional remodeling of a number of distinct tissue types of the animal, including its nervous system. The transcription factor (TF) effectors of 3 different hormonal signaling systems, the insulin-responsive DAF-16/FoxO TF, the TGFß-responsive DAF-3/SMAD TF, and the steroid nuclear hormone receptor, DAF-12/VDR, a homolog of the vitamin D receptor (VDR), were previously shown to be required for entering the dauer arrest stage, but their cellular and temporal focus of action for the underlying cellular remodeling processes remained incompletely understood. Through the generation of conditional alleles that allowed us to spatially and temporally control gene activity, we show here that all 3 TFs are not only required to initiate tissue remodeling upon entry into the dauer stage, as shown before, but are also continuously required to maintain the remodeled state. We show that DAF-3/SMAD is required in sensory neurons to promote and then maintain animal-wide tissue remodeling events. In contrast, DAF-16/FoxO or DAF-12/VDR act cell-autonomously to control anatomical, molecular, and behavioral remodeling events in specific cell types. Intriguingly, we also uncover non-cell autonomous function of DAF-16/FoxO and DAF-12/VDR in nervous system remodeling, indicating the presence of several insulin-dependent interorgan signaling axes. Our findings provide novel perspectives into how hormonal systems control tissue remodeling.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Comunicación Celular/genética , Plasticidad de la Célula/genética , Factores de Transcripción Forkhead/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Animales , Animales Modificados Genéticamente , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/genética , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Sistema Nervioso/crecimiento & desarrollo , Sistema Nervioso/metabolismo , Especificidad de Órganos/genética , Organogénesis/genética , Comunicación Paracrina/genética , Receptores de Calcitriol/genética , Receptores de Calcitriol/fisiología , Receptores Citoplasmáticos y Nucleares/genética , Células Receptoras Sensoriales/metabolismo , Células Receptoras Sensoriales/fisiología , Transducción de Señal/genética
8.
Cell ; 139(6): 1051-3, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005799

RESUMEN

Conventional wisdom holds that the ovary and testis are terminally differentiated organs in adult mammals. However, Uhlenhaut et al. (2009) now report that deletion of a single gene, Foxl2, is sufficient to induce transdifferentiation of ovary into testis in adult mice, suggesting that testicular development is actively repressed throughout the life of females.


Asunto(s)
Transdiferenciación Celular , Ovario/fisiología , Testículo/fisiología , Animales , Línea Celular , Femenino , Proteína Forkhead Box L2 , Factores de Transcripción Forkhead/fisiología , Humanos , Masculino , Ratones , Ovario/citología , Testículo/citología
9.
PLoS Genet ; 17(11): e1009854, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34723967

RESUMEN

The forkhead box (Fox) family of transcription factors are highly conserved and play essential roles in a wide range of cellular and developmental processes. We report an individual with severe neurological symptoms including postnatal microcephaly, progressive brain atrophy and global developmental delay associated with a de novo missense variant (M280L) in the FOXR1 gene. At the protein level, M280L impaired FOXR1 expression and induced a nuclear aggregate phenotype due to protein misfolding and proteolysis. RNAseq and pathway analysis showed that FOXR1 acts as a transcriptional activator and repressor with central roles in heat shock response, chaperone cofactor-dependent protein refolding and cellular response to stress pathways. Indeed, FOXR1 expression is increased in response to cellular stress, a process in which it directly controls HSPA6, HSPA1A and DHRS2 transcripts. The M280L mutant compromises FOXR1's ability to respond to stress, in part due to impaired regulation of downstream target genes that are involved in the stress response pathway. Quantitative PCR of mouse embryo tissues show Foxr1 expression in the embryonic brain. Using CRISPR/Cas9 gene editing, we found that deletion of mouse Foxr1 leads to a severe survival deficit while surviving newborn Foxr1 knockout mice have reduced body weight. Further examination of newborn Foxr1 knockout brains revealed a decrease in cortical thickness and enlarged ventricles compared to littermate wild-type mice, suggesting that loss of Foxr1 leads to atypical brain development. Combined, these results suggest FOXR1 plays a role in cellular stress response pathways and is necessary for normal brain development.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Factores de Transcripción Forkhead/fisiología , Estrés Fisiológico , Animales , Femenino , Factores de Transcripción Forkhead/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Noqueados , Mutación Missense , Fenotipo
10.
PLoS Genet ; 17(11): e1009881, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34780472

RESUMEN

Many tissue-specific stem cells maintain the ability to produce multiple cell types during long periods of non-division, or quiescence. FOXO transcription factors promote quiescence and stem cell maintenance, but the mechanisms by which FOXO proteins promote multipotency during quiescence are still emerging. The single FOXO ortholog in C. elegans, daf-16, promotes entry into a quiescent and stress-resistant larval stage called dauer in response to adverse environmental cues. During dauer, stem and progenitor cells maintain or re-establish multipotency to allow normal development to resume after dauer. We find that during dauer, daf-16/FOXO prevents epidermal stem cells (seam cells) from prematurely adopting differentiated, adult characteristics. In particular, dauer larvae that lack daf-16 misexpress collagens that are normally adult-enriched. Using col-19p::gfp as an adult cell fate marker, we find that all major daf-16 isoforms contribute to opposing col-19p::gfp expression during dauer. By contrast, daf-16(0) larvae that undergo non-dauer development do not misexpress col-19p::gfp. Adult cell fate and the timing of col-19p::gfp expression are regulated by the heterochronic gene network, including lin-41 and lin-29. lin-41 encodes an RNA-binding protein orthologous to LIN41/TRIM71 in mammals, and lin-29 encodes a conserved zinc finger transcription factor. In non-dauer development, lin-41 opposes adult cell fate by inhibiting the translation of lin-29, which directly activates col-19 transcription and promotes adult cell fate. We find that during dauer, lin-41 blocks col-19p::gfp expression, but surprisingly, lin-29 is not required in this context. Additionally, daf-16 promotes the expression of lin-41 in dauer larvae. The col-19p::gfp misexpression phenotype observed in dauer larvae with reduced daf-16 requires the downregulation of lin-41, but does not require lin-29. Taken together, this work demonstrates a novel role for daf-16/FOXO as a heterochronic gene that promotes expression of lin-41/TRIM71 to contribute to multipotent cell fate in a quiescent stem cell model.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/citología , Linaje de la Célula , Factores de Transcripción Forkhead/fisiología , Factores de Transcripción/fisiología , Animales , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas de Caenorhabditis elegans/genética , Colágeno/metabolismo , Factores de Transcripción Forkhead/genética , Larva/citología , Larva/metabolismo , Factores de Transcripción/genética
11.
EMBO J ; 38(9)2019 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-30886050

RESUMEN

Regulatory T cells (Tregs) are crucial mediators of immune control. The characteristic gene expression and suppressive functions of Tregs depend considerably on the stable expression and activity of the transcription factor FOXP3. Transcriptional regulation of the Foxp3 gene has been studied in depth, but both the expression and function of this factor are also modulated at the protein level. However, the molecular players involved in posttranslational FOXP3 regulation are just beginning to be elucidated. Here, we found that TRAF6-deficient Tregs were dysfunctional in vivo; mice with Treg-restricted deletion of TRAF6 were resistant to implanted tumors and displayed enhanced anti-tumor immunity. We further determined that FOXP3 undergoes K63-linked ubiquitination at lysine 262 mediated by the E3 ligase TRAF6. In the absence of TRAF6 activity or upon mutation of the ubiquitination site, FOXP3 displayed aberrant, perinuclear accumulation and disrupted regulatory function. Thus, K63-linked ubiquitination by TRAF6 ensures proper localization of FOXP3 and facilitates the transcription factor's gene-regulating activity in Tregs. These results implicate TRAF6 as a key posttranslational, Treg-stabilizing regulator that may be targeted in novel tolerance-breaking therapies.


Asunto(s)
Colitis/inmunología , Factores de Transcripción Forkhead/fisiología , Lisina/metabolismo , Melanoma Experimental/inmunología , Linfocitos T Reguladores/inmunología , Factor 6 Asociado a Receptor de TNF/fisiología , Ubiquitinación , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Colitis/inducido químicamente , Colitis/metabolismo , Colitis/patología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Melanoma Experimental/metabolismo , Melanoma Experimental/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Linfocitos T Reguladores/metabolismo , Linfocitos T Reguladores/patología
12.
Development ; 147(4)2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31988185

RESUMEN

Organogenesis requires precise interactions between a developing tissue and its environment. In vertebrates, the developing eye is surrounded by a complex extracellular matrix as well as multiple mesenchymal cell populations. Disruptions to either the matrix or periocular mesenchyme can cause defects in early eye development, yet in many cases the underlying mechanism is unknown. Here, using multidimensional imaging and computational analyses in zebrafish, we establish that cell movements in the developing optic cup require neural crest. Ultrastructural analysis reveals that basement membrane formation around the developing eye is also dependent on neural crest, but only specifically around the retinal pigment epithelium. Neural crest cells produce the extracellular matrix protein nidogen: impairing nidogen function disrupts eye development, and, strikingly, expression of nidogen in the absence of neural crest partially restores optic cup morphogenesis. These results demonstrate that eye formation is regulated in part by extrinsic control of extracellular matrix assembly.This article has an associated 'The people behind the papers' interview.


Asunto(s)
Membrana Basal/embriología , Ojo/embriología , Cresta Neural/embriología , Alelos , Animales , Sistemas CRISPR-Cas , Proteínas de Unión al Calcio/fisiología , Movimiento Celular , Electroforesis Capilar , Matriz Extracelular/fisiología , Proteínas de la Matriz Extracelular/fisiología , Factores de Transcripción Forkhead/fisiología , Regulación del Desarrollo de la Expresión Génica , Genotipo , Mesodermo/embriología , Microscopía Electrónica de Transmisión , Morfogénesis , Mutación , Cresta Neural/citología , Organogénesis , Retina/embriología , Epitelio Pigmentado de la Retina/embriología , Transducción de Señal , Factor de Transcripción AP-2/fisiología , Pez Cebra , Proteínas de Pez Cebra/fisiología
13.
Proc Natl Acad Sci U S A ; 117(22): 12258-12268, 2020 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-32414925

RESUMEN

Foxp3-expressing regulatory T cells (Tregs) can be generated in vitro by antigenic stimulation of conventional T cells (Tconvs) in the presence of TGF-ß and IL-2. However, unlike Foxp3+ naturally occurring Tregs, such in vitro induced Tregs (iTregs) are functionally unstable mainly because of incomplete Treg-type epigenetic changes at Treg signature genes such as Foxp3 Here we show that deprivation of CD28 costimulatory signal at an early stage of iTreg generation is able to establish Treg-specific DNA hypomethylation at Treg signature genes. It was achieved, for example, by TCR/TGF-ß/IL-2 stimulation of CD28-deficient Tconvs or CD28-intact Tconvs without anti-CD28 agonistic mAb or with CD80/CD86-blocked or -deficient antigen-presenting cells. The signal abrogation could induce Treg-type hypomethylation in memory/effector as well as naive Tconvs, while hindering Tconv differentiation into effector T cells. Among various cytokines and signal activators/inhibitors, TNF-α and PKC agonists inhibited the hypomethylation. Furthermore, CD28 signal deprivation significantly reduced c-Rel expression in iTregs; and the specific genomic perturbation of a NF-κB binding motif at the Foxp3 CNS2 locus enhanced the locus-specific DNA hypomethylation even in CD28 signaling-intact iTregs. In addition, in vitro maintenance of such epigenome-installed iTregs with IL-2 alone, without additional TGF-ß or antigenic stimulation, enabled their expansion and stabilization of Treg-specific DNA hypomethylation. These iTregs indeed stably expressed Foxp3 after in vivo transfer and effectively suppressed antigen-specific immune responses. Taken together, inhibition of the CD28-PKC-NF-κB signaling pathway in iTreg generation enables de novo acquisition of Treg-specific DNA hypomethylation at Treg signature genes and abundant production of functionally stable antigen-specific iTregs for therapeutic purposes.


Asunto(s)
Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/inmunología , Metilación de ADN , Epigénesis Genética , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular , Citocinas/metabolismo , Femenino , Interleucina-6/metabolismo , Masculino , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal , Linfocitos T Reguladores/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
14.
Dev Biol ; 477: 219-231, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34107272

RESUMEN

The endostyle is a ventral pharyngeal organ used for internal filter feeding of basal chordates and is considered homologous to the follicular thyroid of vertebrates. It contains mucus-producing (glandular) and thyroid-equivalent regions organized along the dorsoventral (DV) axis. Although thyroid-related genes (Nkx2-1, FoxE, and thyroid peroxidase (TPO)) are known to be expressed in the endostyle, their roles in establishing regionalization within the organ have not been demonstrated. We report that Nkx2-1 and FoxE are essential for establishing DV axial identity in the endostyle of Oikopleura dioica. Genome and expression analyses showed von Willebrand factor-like (vWFL) and TPO/dual oxidase (Duox)/Nkx2-1/FoxE as orthologs of glandular and thyroid-related genes, respectively. Knockdown experiments showed that Nkx2-1 is necessary for the expression of glandular and thyroid-related genes, whereas FoxE is necessary only for thyroid-related genes. Moreover, Nkx2-1 expression is necessary for FoxE expression in larvae during organogenesis. The results demonstrate the essential roles of Nkx2-1 and FoxE in establishing regionalization in the endostyle, including (1) the Nkx2-1-dependent glandular region, and (2) the Nkx2-1/FoxE-dependent thyroid-equivalent region. DV axial regionalization may be responsible for organizing glandular and thyroid-equivalent traits of the pharynx along the DV axis.


Asunto(s)
Factores de Transcripción Forkhead/fisiología , Hormonas Tiroideas/fisiología , Factor Nuclear Tiroideo 1/fisiología , Urocordados/embriología , Animales , Moco , Glándula Tiroides/embriología , Glándula Tiroides/fisiología , Urocordados/anatomía & histología , Urocordados/fisiología
15.
Clin Immunol ; 234: 108914, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34954131

RESUMEN

Increasing fine particulate matter (PM2.5) and epigenetic modifications are closely associated with the pathogenesis of asthma, but the definite mechanism remains unclear. The traffic-related PM2.5 exposure aggravated pulmonary inflammation and changed the methylation level of interferon gamma (Ifng) and interleukin (Il)4 genes, and then altered levels of affiliated cytokines of IFN-γ and IL-4 in rats with allergic airway inflammation. It also increased the level of miR146a and decreased the level of miR31. In addition, transcription factors of nuclear factor kappa B (NF-κB) and signal transducer and activator of transcription 6 (Stat6) rose; forkhead box P3 (Foxp3) and signal transducer and activator of transcription 4 (Stat4) lowered. The traffic-related PM2.5 altered epigenetic modifications in allergic airway inflammation of rats leading to inflammation exacerbation through impaired regulatory T (Treg) cells function and T-helper type 1 (Th1)/Th2 cells imbalance, which provided a new target for the treatment and control of asthma.


Asunto(s)
Asma/etiología , Metilación de ADN , Material Particulado/toxicidad , Emisiones de Vehículos/toxicidad , Animales , Asma/genética , Asma/inmunología , Citocinas/análisis , Factores de Transcripción Forkhead/fisiología , Interferón gamma/genética , Interferón gamma/fisiología , Interleucina-4/genética , Interleucina-4/fisiología , Masculino , MicroARNs/análisis , FN-kappa B/fisiología , Ratas , Ratas Sprague-Dawley , Factores de Transcripción STAT/análisis , Linfocitos T Reguladores/inmunología
16.
Am J Hum Genet ; 105(3): 549-561, 2019 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-31447097

RESUMEN

FOXN1 is the master regulatory gene of thymic epithelium development. FOXN1 deficiency leads to thymic aplasia, alopecia, and nail dystrophy, accounting for the nude/severe combined immunodeficiency (nu/SCID) phenotype in humans and mice. We identified several newborns with low levels of T cell receptor excision circles (TRECs) and T cell lymphopenia at birth, who carried heterozygous loss-of-function FOXN1 variants. Longitudinal analysis showed persistent T cell lymphopenia during infancy, often associated with nail dystrophy. Adult individuals with heterozygous FOXN1 variants had in most cases normal CD4+ but lower than normal CD8+ cell counts. We hypothesized a FOXN1 gene dosage effect on the function of thymic epithelial cells (TECs) and thymopoiesis and postulated that these effects would be more prominent early in life. To test this hypothesis, we analyzed TEC subset frequency and phenotype, early thymic progenitor (ETP) cell count, and expression of FOXN1 target genes (Ccl25, Cxcl12, Dll4, Scf, Psmb11, Prss16, and Cd83) in Foxn1nu/+ (nu/+) mice and age-matched wild-type (+/+) littermate controls. Both the frequency and the absolute count of ETP were significantly reduced in nu/+ mice up to 3 weeks of age. Analysis of the TEC compartment showed reduced expression of FOXN1 target genes and delayed maturation of the medullary TEC compartment in nu/+ mice. These observations establish a FOXN1 gene dosage effect on thymic function and identify FOXN1 haploinsufficiency as an important genetic determinant of T cell lymphopenia at birth.


Asunto(s)
Factores de Transcripción Forkhead/genética , Heterocigoto , Linfopenia/genética , Linfocitos T/metabolismo , Timo/citología , Adulto , Anciano , Animales , Preescolar , Femenino , Factores de Transcripción Forkhead/fisiología , Humanos , Lactante , Recién Nacido , Masculino , Ratones , Ratones SCID , Persona de Mediana Edad , Adulto Joven
17.
Immunity ; 39(5): 949-62, 2013 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-24238343

RESUMEN

Stable Foxp3 expression is crucial for regulatory T (Treg) cell function. We observed that antigen-driven activation and inflammation in the CNS promoted Foxp3 instability selectively in the autoreactive Treg cells that expressed high amounts of Foxp3 before experimental autoimmune encephalitis induction. Treg cells with a demethylated Treg-cell-specific demethylated region in the Foxp3 locus downregulated Foxp3 transcription in the inflamed CNS during the induction phase of the response. Stable Foxp3 expression returned at the population level with the resolution of inflammation or was rescued by IL-2-anti-IL-2 complex treatment during the antigen priming phase. Thus, a subset of fully committed self-antigen-specific Treg cells lost Foxp3 expression during an inflammatory autoimmune response and might be involved in inadequate control of autoimmunity. These results have important implications for Treg cell therapies and give insights into the dynamics of the Treg cell network during autoreactive CD4(+) T cell effector responses in vivo.


Asunto(s)
Autoantígenos/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Factores de Transcripción Forkhead/fisiología , Regulación de la Expresión Génica/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linaje de la Célula , Sistema Nervioso Central/inmunología , Metilación de ADN , Regulación hacia Abajo/inmunología , Factores de Transcripción Forkhead/biosíntesis , Factores de Transcripción Forkhead/genética , Genes Reporteros , Ganglios Linfáticos/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Glicoproteína Mielina-Oligodendrócito/inmunología , Fragmentos de Péptidos/inmunología , Receptores de Interleucina-2/fisiología , Proteínas Recombinantes de Fusión/inmunología , Secuencias Reguladoras de Ácidos Nucleicos , Organismos Libres de Patógenos Específicos
18.
Malar J ; 21(1): 32, 2022 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-35109868

RESUMEN

BACKGROUND: Regulatory T cells are known to play a key role to counter balance the protective immune response and immune mediated pathology. However, the role of naturally occurring regulatory cells CD4+CD25+Foxp3+ in malaria infection during the disease pathogenesis is controversial. Beside this, ICOS molecule has been shown to be involved in the development and function of regulatory T cell enhance IL-10 production. Therefore, possible involvement of the ICOS dependent regulatory CD4+ICOS+Foxp3+ T cells in resistance/susceptibility during malaria parasite is explored in this study. METHODS: 5 × 105 red blood cells infected with non-lethal and lethal parasites were inoculated in female Balb/c mice by intra-peritoneal injection. Infected or uninfected mice were sacrificed at early (3rd day post infection) and later stage (10th day post infection) of infection. Harvested cells were analysed by using flow cytometer and serum cytokine by Bioplex assay. RESULTS: Thin blood films show that percentages of parasitaemia increases with disease progression in infections with the lethal malaria parasite and mice eventually die by day 14th post-infection. Whereas in case of non-lethal malaria parasite, parasitaemia goes down by 7th day post infection and gets cleared within 13th day. The number of CD4+ ICOS+ T cells increases in lethal infection with disease progression. Surprisingly, in non-lethal parasite, ICOS expression decreases after day 7th post infection as parasitaemia goes down. The frequency of CD4+ICOS+FoxP3+ Tregs was significantly higher in lethal parasitic infection as compared to the non-lethal parasite. The level of IL-12 cytokine was remarkably higher in non-lethal infection compared to the lethal infection. In contrast, the level of IL-10 cytokines was higher in lethal parasite infection compared to the non-lethal parasite. CONCLUSION: Taken together, these data suggest that lethal parasite induce immunosuppressive environment, protecting from host immune responses and help the parasite to survive whereas non-lethal parasite leads to low frequencies of Treg cells seldom impede immune response that allow the parasite to get self-resolved.


Asunto(s)
Malaria/etiología , Linfocitos T Reguladores/fisiología , Animales , Antígenos CD4/fisiología , Citocinas/análisis , Femenino , Citometría de Flujo , Factores de Transcripción Forkhead/fisiología , Humanos , Proteína Coestimuladora de Linfocitos T Inducibles/fisiología , Interleucina-10/análisis , Malaria/diagnóstico , Malaria/inmunología , Ratones , Ratones Endogámicos BALB C , Parasitemia/diagnóstico , Parasitemia/parasitología , Fragmentos de Péptidos/fisiología , Plasmodium berghei , Plasmodium chabaudi , Plasmodium yoelii , Organismos Libres de Patógenos Específicos , Bazo/citología
19.
Exp Cell Res ; 399(1): 112422, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33338479

RESUMEN

PURPOSE: Epithelial ovarian cancer (EOC) is a highly fatal gynecological cancer. A long noncoding RNA (lncRNA) gastric cancer-associated lncRNA1 (GClnc1) has been revealed to play critical roles in metastasis. Therefore, the present study aims to explore the correlation between GClnc1 and the metastasis and progression of EOC. METHODS: First, 57 paired EOC and paracancerous tissues were collected to detect GClnc1 expression by RT-qPCR. Subsequently, OVC1 and SKOV3 cells with GClnc1 silencing/overexpression were developed to detect changes in cell activity, apoptosis, migration and invasion abilities. Then, the subcellular localization of GClnc1 was detected by nuclear/cytoplasmic fractionation, ISH and FISH assays. The binding relationships between GClnc1 and forkhead box protein C2 (FOXC2), and between FOXC2 and NOTCH1 were predicted and verified. RESULTS: GClnc1 was significantly overexpressed in EOC tissues, and knockdown of GClnc1 inhibited cell viability and promoted apoptosis. Moreover, GClnc1 in the nucleus bound to the transcription factor FOXC2, thereby activating the transcription of NOTCH1. NOTCH1 overexpression enhanced the proliferation and epithelial-mesenchymal transition of SKOV3 and OVC1 cells. Moreover, NOTCH1 activated the NF-κB/Snail signaling. Finally, in vivo experiments demonstrated that GClnc1 knockdown suppressed the growth and metastasis of SKOV3 and OVC1 cells in vivo. CONCLUSIONS: GClnc1 promoted NOTCH1 transcription by recruiting FOXC2, thereby activating the NF-κB/Snail signaling and promoting EOC cell growth and metastasis.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Factores de Transcripción Forkhead/fisiología , Neoplasias Ováricas/patología , ARN Largo no Codificante/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma Epitelial de Ovario/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Factores de Transcripción Forkhead/metabolismo , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Ligamiento Genético , Humanos , Persona de Mediana Edad , FN-kappa B/genética , FN-kappa B/metabolismo , Neoplasias Ováricas/genética , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transducción de Señal/genética , Factores de Transcripción de la Familia Snail/genética , Factores de Transcripción de la Familia Snail/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
20.
Hum Mol Genet ; 28(8): 1286-1297, 2019 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-30561639

RESUMEN

Molecular mechanisms governing the development of the human cochlea remain largely unknown. Through genome sequencing, we identified a homozygous FOXF2 variant c.325A>T (p.I109F) in a child with profound sensorineural hearing loss (SNHL) associated with incomplete partition type I anomaly of the cochlea. This variant is not found in public databases or in over 1000 ethnicity-matched control individuals. I109 is a highly conserved residue in the forkhead box (Fox) domain of FOXF2, a member of the Fox protein family of transcription factors that regulate the expression of genes involved in embryogenic development as well as adult life. Our in vitro studies show that the half-life of mutant FOXF2 is reduced compared to that of wild type. Foxf2 is expressed in the cochlea of developing and adult mice. The mouse knockout of Foxf2 shows shortened and malformed cochleae, in addition to altered shape of hair cells with innervation and planar cell polarity defects. Expressions of Eya1 and Pax3, genes essential for cochlear development, are reduced in the cochleae of Foxf2 knockout mice. We conclude that FOXF2 plays a major role in cochlear development and its dysfunction leads to SNHL and developmental anomalies of the cochlea in humans and mice.


Asunto(s)
Cóclea/embriología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/fisiología , Adulto , Animales , Niño , Cóclea/metabolismo , Cóclea/fisiología , Desarrollo Embrionario , Femenino , Células Ciliadas Auditivas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Nucleares/genética , Proteínas Nucleares/fisiología , Organogénesis , Factor de Transcripción PAX3/genética , Factor de Transcripción PAX3/fisiología , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/fisiología , Transducción de Señal/genética , Secuenciación Completa del Genoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA