Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 348
Filtrar
1.
Cell Rep ; 37(6): 109961, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758310

RESUMEN

Following infection or immunization, memory B cells (MBCs) and long-lived plasma cells provide humoral immunity that can last for decades. Most principles of MBC biology have been determined with hapten-protein carrier models or fluorescent protein immunizations. Here, we examine the temporal dynamics of the germinal center (GC) B cell and MBC response following mouse influenza A virus infection. We find that antiviral B cell responses within the lung-draining mediastinal lymph node (mLN) and the spleen are distinct in regard to duration, enrichment for antigen-binding cells, and class switching dynamics. While splenic GCs dissolve after 6 weeks post-infection, mLN hemagglutinin-specific (HA+) GCs can persist for 22 weeks. Persistent GCs continuously differentiate MBCs, with "peak" and "late" GCs contributing equal numbers of HA+ MBCs to the long-lived compartment. Our findings highlight critical aspects of persistent GC responses and MBC differentiation following respiratory virus infection with direct implications for developing effective vaccination strategies.


Asunto(s)
Anticuerpos Antivirales/inmunología , Centro Germinal/inmunología , Memoria Inmunológica , Virus de la Influenza A/fisiología , Células B de Memoria/inmunología , Infecciones por Orthomyxoviridae/inmunología , Proteínas de Dominio T Box/fisiología , Animales , Diferenciación Celular , Femenino , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología
2.
Mol Genet Genomics ; 296(4): 809-821, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33866394

RESUMEN

Holt-Oram syndrome (HOS) is a rare disorder characterized by cardiac and upper-limb defects. Pathogenic variants in TBX5-a gene encoding a transcription factor important for heart and skeletal development-are the only known cause of HOS. Here, we present the identification and functional analysis of two novel TBX5 pathogenic variants found in two individuals with HOS presenting distinct phenotypes. The individual with the c.905delA variant has a severe cardiac phenotype but mild skeletal defects, unlike the individual with the c.246_249delGATG variant who has no cardiac problems but severe upper limbs malformations, including phocomelia. Both frameshift variants, c.246_249delGATG and c.905delA, generate mRNAs harbouring premature stop codons which, if not degraded by nonsense mediated decay, will lead to the production of shorter TBX5 proteins, p.Gln302Argfs*92 and p.Met83Phefs*6, respectively. Immunocytochemistry results suggest that both mutated proteins are produced and furthermore, like the wild-type protein, p.Gln302Argfs*92 mutant appears to be mainly localized in the nucleus, in contrast with p.Met83Phefs*6 mutant that displays a higher level of cytoplasmic localization. In addition, luciferase activity analysis revealed that none of the TBX5 mutants are capable of transactivating the NPPA promoter. In conclusion, our results provide evidence that both pathogenic variants cause a severe TBX5 loss-of-function, dramatically reducing its biological activity. The absence of cardiac problems in the individual with the p.Met83Phefs*6 variant supports the existence of other mechanisms/genes underlying the pathogenesis of HOS and/or the existence of an age-related delay in the development of a more serious cardiac phenotype. Further studies are required to understand the differential effects observed in the phenotypes of both individuals.


Asunto(s)
Anomalías Múltiples/genética , Anomalías Múltiples/patología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Defectos del Tabique Interatrial/genética , Defectos del Tabique Interatrial/patología , Deformidades Congénitas de las Extremidades Inferiores/genética , Deformidades Congénitas de las Extremidades Inferiores/patología , Proteínas de Dominio T Box/genética , Deformidades Congénitas de las Extremidades Superiores/genética , Deformidades Congénitas de las Extremidades Superiores/patología , Adulto , Anciano de 80 o más Años , Células Cultivadas , Análisis Citogenético , Análisis Mutacional de ADN , Estudios de Asociación Genética , Heterogeneidad Genética , Células HEK293 , Humanos , Masculino , Mutación/fisiología , Fenotipo , Proteínas de Dominio T Box/fisiología
3.
Gene ; 785: 145602, 2021 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-33766712

RESUMEN

T-box transcription factor 3 (TBX3) gene encodes a transcriptional suppressor and plays an important role in embryonic development, which belongs to the T-box family. TBX3 also has been found to be associated with body size traits in horse that is a relative of donkey. Therefore, TBX3 is considered as a promising candidate gene for economic traits of donkey. This study aimed to reveal the significant variation of TBX3 gene in Dezhou donkey and explores the relationship between genotypes and body sizes. In this study, an A > G mutation was found in the intron 2 of TBX3 gene by sequencing, and three genotypes (AA, GG and AG) were identified in 380 Dezhou donkey individuals with Tm-shift method. Association analysis illustrated that there were significant differences between AA and GG genotype in body length, body height, chest depth, chest circumference, body weight, hucklebone width and rump length. Our results demonstrated that the polymorphism of TBX3 is significantly associated with body size traits, which can serve as a marker to improve donkey production performance.


Asunto(s)
Tamaño Corporal/genética , Equidae/anatomía & histología , Equidae/genética , Proteínas de Dominio T Box/genética , Animales , Femenino , Genotipo , Intrones/genética , Masculino , Mutación , Polimorfismo de Nucleótido Simple , Proteínas de Dominio T Box/fisiología
4.
Leukemia ; 35(11): 3152-3162, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-33731848

RESUMEN

Genome-wide association studies identified a single-nucleotide polymorphism (SNP) affecting the transcription factor Eomesodermin (EOMES) associated with a significantly increased risk to develop chronic lymphocytic leukemia (CLL). Epigenetic analyses, RNA sequencing, and flow cytometry revealed that EOMES is not expressed in CLL cells, but in CD8+ T cells for which EOMES is a known master regulator. We thus hypothesized that the increased CLL risk associated with the EOMES SNP might be explained by its negative impact on CD8+ T-cell-mediated immune control of CLL. Flow cytometry analyses revealed a higher EOMES expression in CD8+ T cells of CLL patients compared to healthy individuals, and an accumulation of PD-1+ EOMES+ CD8+ T cells in lymph nodes rather than blood or bone marrow in CLL. This was in line with an observed expansion of EOMES+ CD8+ T cells in the spleen of leukemic Eµ-TCL1 mice. As EOMES expression was highest in CD8+ T cells that express inhibitory receptors, an involvement of EOMES in T-cell exhaustion and dysfunction seems likely. Interestingly, Eomes-deficiency in CD8+ T cells resulted in their impaired expansion associated with decreased CLL control in mice. Overall, these observations suggest that EOMES is essential for CD8+ T-cell expansion and/or maintenance, and therefore involved in adaptive immune control of CLL.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Leucemia Linfocítica Crónica de Células B/inmunología , Ganglios Linfáticos/inmunología , Proteínas de Dominio T Box/metabolismo , Proteínas de Dominio T Box/fisiología , Animales , Estudios de Casos y Controles , Femenino , Estudio de Asociación del Genoma Completo , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas de Dominio T Box/genética
5.
Nat Cell Biol ; 23(1): 61-74, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33420489

RESUMEN

Extra-embryonic mesoderm (ExM)-composed of the earliest cells that traverse the primitive streak-gives rise to the endothelium as well as haematopoietic progenitors in the developing yolk sac. How a specific subset of ExM becomes committed to a haematopoietic fate remains unclear. Here we demonstrate using an embryonic stem cell model that transient expression of the T-box transcription factor Eomesodermin (Eomes) governs haemogenic competency of ExM. Eomes regulates the accessibility of enhancers that the transcription factor stem cell leukaemia (SCL) normally utilizes to specify primitive erythrocytes and is essential for the normal development of Runx1+ haemogenic endothelium. Single-cell RNA sequencing suggests that Eomes loss of function profoundly blocks the formation of blood progenitors but not specification of Flk-1+ haematoendothelial progenitors. Our findings place Eomes at the top of the transcriptional hierarchy regulating early blood formation and suggest that haemogenic competence is endowed earlier during embryonic development than was previously appreciated.


Asunto(s)
Células Madre Embrionarias/citología , Hemangioblastos/citología , Mesodermo/citología , Proteínas de Dominio T Box/fisiología , Saco Vitelino/citología , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Células Madre Embrionarias/metabolismo , Femenino , Hemangioblastos/metabolismo , Masculino , Mesodermo/metabolismo , Ratones Noqueados , Embarazo , RNA-Seq , Análisis de la Célula Individual , Proteína 1 de la Leucemia Linfocítica T Aguda/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Saco Vitelino/metabolismo
6.
Insect Biochem Mol Biol ; 129: 103516, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33412239

RESUMEN

The T-box genes are essential transcription factors during limb development. In Drosophila, Dorsocross (Doc) and optomotor-blind (omb), members of the Tbx2 and Tbx6 families, are best studied in the Drosophila wing development. Despite prominently expressed in leg discs, the specific function of these genes in leg growth is still not revealed. Here we demonstrated that Doc and omb regulated the morphogenesis of leg intermediate regions in a functionally redundant manner. Loss of Doc or omb individually did not result in any developmental defects of the legs, but loss of both genes induced significant defects in femur and proximal tibia of the adult legs. These genes located in the dorsal domain, where the Doc region expanded and cross-overlapped with the omb region corresponding to the presumptive leg intermediate region. We detected that the normal epithelial folds in the leg discs were disrupted along with dorsal repression of cell proliferation and activation of cell apoptosis when Doc and omb were both reduced. Furthermore, the dorsal expression of dachshund (dac), a canonical leg developmental gene specifying the leg intermediate region, was maintained by Doc and omb. Meanwhile, the Notch pathway was compromised in the dorsal domain when these genes were reduced, which might contribute to the joint defect of the adult leg intermediate regions. Our study provides cytological and genetic evidence for understanding the redundant function of Doc and omb in leg morphogenesis.


Asunto(s)
Proteínas de Drosophila/fisiología , Drosophila/crecimiento & desarrollo , Extremidad Inferior/crecimiento & desarrollo , Proteínas del Tejido Nervioso/fisiología , Proteínas de Dominio T Box/fisiología , Factores de Transcripción/fisiología , Animales , Drosophila/metabolismo , Proteínas de Drosophila/metabolismo , Proteínas Nucleares/metabolismo , Receptores Notch/metabolismo
7.
J Invest Dermatol ; 141(5): 1274-1285.e5, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33068596

RESUMEN

T-bet and signal transducer and activator of transcription (STAT) 6 are critical factors for helper T-cell differentiation in humans and mice. Additionally, polymorphisms in TBX21 (T-bet) and STAT6 are associated with the susceptibility of allergic diseases. However, precise mechanisms of the reciprocal regulation between T-bet and STAT6 in allergy remain unclear. To determine the reciprocal regulation in vivo, we investigated the phenotype of T-bet/STAT6 double-deficient (T-bet-/- STAT6-/-) mice. Unexpectedly, T-bet-/- STAT6-/- mice but not T-bet-/- mice or STAT6-/- mice spontaneously developed severe dermatitis. Not only eosinophils and mast cells but also CD4+ T cells infiltrated into the skin of T-bet-/- STAT6-/- mice. Adoptive transfer of CD4+ T cells of T-bet-/- STAT6-/- mice into severe combined immunodeficient mice induced the accumulation of eosinophils and mast cells in the skin, whereas depletion of CD4+ T cells ameliorated the dermatitis in T-bet-/- STAT6-/- mice. Comprehensive transcriptome analyses revealed that IL-9 expression was enhanced in T-bet-/- STAT6-/- CD4+ T cells. Indeed, IL-9 neutralization ameliorated the dermatitis in T-bet-/- STAT6-/- mice. T-bet-/- STAT6-/- CD4+ T cells expressed functional thymic stromal lymphopoietin receptors and produced large amounts of IL-9 on thymic stromal lymphopoietin stimulation. These results indicate that T-bet and STAT6 coordinately suppress atopic dermatitis-like skin inflammation, possibly by inhibiting thymic stromal lymphopoietin-dependent IL-9 production in CD4+ T cells.


Asunto(s)
Dermatitis Atópica/prevención & control , Interleucina-9/fisiología , Factor de Transcripción STAT6/fisiología , Proteínas de Dominio T Box/fisiología , Animales , Linfocitos T CD4-Positivos/inmunología , Citocinas/fisiología , Ratones , Ratones Endogámicos BALB C , Linfopoyetina del Estroma Tímico
8.
Front Immunol ; 11: 2125, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32983171

RESUMEN

Although CD4+ T cell memory is a critical component of adaptive immunity, antigen-specific CD4+ T cell recall responses to secondary infection have been inadequately studied. Here we examine the kinetics of the secondary response in an important immunological model, infection with attenuated Listeria monocytogenes (Lm). We identify CD4+ T cell subsets that preferentially expand during a secondary response and highlight the importance of prime-boost strategies in expanding and maintaining antigen-specific, tissue-resident memory CD4+ T cells. Following intravenous infection with an attenuated strain of Lm, we found that total antigen-specific CD4+ T cells responded more robustly in secondary compared with primary infection, reaching near-peak levels in secondary lymphoid organs (SLOs) and the liver by three days post-infection. During the secondary response, CD4+ T cells also contracted more quickly. Primary Lm infection generated two main classes of effector cells: Th1 cells that assist macrophages and T follicular helper (Tfh) cells that aid B cells in antibody production. We found that during the secondary response, a population of Ly6C+ Tfh cells emerged in SLOs and was the basis for the skewing of this response to a Tfh phenotype. Deletion of T-bet in T cells precluded development of Ly6C+ Tfh cells, but did not alter anti-Lm antibody responses. Moreover, during recall responses, CD49a+ Th1 cells preferentially expanded and accumulated in the liver, achieving a new set point. Parabiosis experiments indicated that, in contrast to Tfh cells and most splenic Th1 cells, the majority of CD49a+ Th1 cells in the liver were tissue resident. Overall, these data demonstrate a robust secondary CD4+ T cell response that differs in kinetics and composition from the primary response and provide insight into targets to enhance both peripheral and tissue-resident CD4+ T cell responses.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Memoria Inmunológica , Listeriosis/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Epítopos , Inmunofenotipificación , Cinética , Listeria monocytogenes , Hígado/inmunología , Activación de Linfocitos , Tejido Linfoide/inmunología , Ratones , Ratones Endogámicos C57BL , Parabiosis , Organismos Libres de Patógenos Específicos , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/fisiología , Células TH1/inmunología
9.
Mol Cell Endocrinol ; 518: 111037, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-32966862

RESUMEN

Ubiquitin-specific protease 7 (USP7/HAUSP) is known to regulate multiple cellular phenomena, including cell cycle progression and proliferation, and is involved in binding and stabilizing specific target proteins through deubiquitylation. However, the detailed role of USP7 in papillary thyroid carcinoma (PTC) remains to be investigated. In this study, our results showed that USP7 was upregulated in PTC tissues compared with adjacent nontumour tissues. Consistently, a series of gain/loss functional assays in vivo and in vitro demonstrated the role of USP7 in promoting PTC cell proliferation. Furthermore, we showed that there was a negative correlation between USP7 and the CDK inhibitor p57KIP2 expression in PTC tissues and that USP7 facilitated PTC cell proliferation by inhibiting p57KIP2. Mechanistically, USP7 inhibited p57KIP2 expression by modulating TBX3, directly binding to TBX3, and decreasing its ubiquitination and degradation. Our findings demonstrated that USP7 played a critical oncogenic role in PTC tumorigenesis, suggesting that USP7 might act as a prognostic and therapeutic target for PTC progression.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Proteínas de Dominio T Box/fisiología , Cáncer Papilar Tiroideo/patología , Neoplasias de la Tiroides/patología , Peptidasa Específica de Ubiquitina 7/fisiología , Proliferación Celular/genética , Células Cultivadas , Humanos , Interferencia de ARN , Transducción de Señal/genética , Proteínas de Dominio T Box/genética , Cáncer Papilar Tiroideo/genética , Neoplasias de la Tiroides/genética
10.
FASEB J ; 34(11): 15062-15079, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32951265

RESUMEN

The transcription factor TBX1 is the major gene implicated in 22q11.2 deletion syndrome (22q11.2DS). The complex clinical phenotype includes vascular anomalies and a recent report presented new cases of primary lymphedema in 22q11.2DS patients. We have previously shown that TBX1 is required for systemic lymphatic vessel development in prenatal mice and it is critical for their survival postnatally. Using loss-of-function genetics and transgenesis in the mouse, we show here a strong genetic interaction between Tbx1 and Vegfr3 in cardiac lymphangiogenesis. Intriguingly, we found that different aspects of the cardiac lymphatic phenotype in Tbx1-Vegfr3 compound heterozygotes were regulated independently by the two genes, with Tbx1 primarily regulating vessel numbers and Vegfr3 vessel morphology. Consistent with this observation, Tbx1Cre -activated expression of a Vegfr3 transgene rescued partially the cardiac lymphatic abnormalities in compound heterozygotes. Through time-controlled genetic experiments, we show that Tbx1 is activated and required in cardiac lymphatic endothelial cell (LEC) progenitors between E10.5 and E11.5. Furthermore, we found that it is also required later in development for the growth of the cardiac lymphatics. Finally, our study revealed a differential sensitivity between ventral and dorsal cardiac lymphatics to the effects of altered Tbx1 and Vegfr3 gene dosage, and we show that this likely results from an earlier requirement for Tbx1 in ventral cardiac LEC progenitors.


Asunto(s)
Corazón/fisiopatología , Linfangiogénesis , Vasos Linfáticos/patología , Células Madre Embrionarias de Ratones/patología , Proteínas de Dominio T Box/fisiología , Receptor 3 de Factores de Crecimiento Endotelial Vascular/fisiología , Animales , Femenino , Heterocigoto , Vasos Linfáticos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Madre Embrionarias de Ratones/metabolismo
11.
Mol Med Rep ; 22(2): 1145-1154, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32627000

RESUMEN

The increasing burden of diabetes in low and middle­income countries is attributable to both genetic and epigenetic factors. Environmental­ and lifestyle­associated changes are also considered to be important contributors to this disease. The resultant co­morbidities arising from micro­and macrovascular changes in diabetes are difficult to manage and are an economic burden. However, very little is known about the molecular mechanisms that drive this phenotype. The present study aimed to investigate the role of sirtuin 1 (SIRT1)­ and transcription box­3 (TBX­3)­mediated regulation of endothelial dysfunction, given the significance of SIRT1 in glucose metabolism and the role of TBX­3 in the maintenance of cellular proliferation, senescence and apoptosis. Following the recruitment of adult patients with and without diabetes, both SIRT1 and TBX­3 expression was confirmed to be present in the sera of the patients with diabetes and the patients without diabetes; however, both SIRT1 and TBX­3 expression levels were higher in the sera of the patients with diabetes. Human umbilical vein endothelial cells (HUVECs) were further used for in vitro studies. Using TBX­3 and SIRT1 knockdown models, the cellular responses to proliferation, migration, invasion and tube formation were investigated using an MTS, cell cycle analysis, wound healing, Transwell and tube formation assay, respectively. Western blotting was also used to determine the downstream signaling pathways involved. The genetic knockdown of TBX­3 in hyperglycemic conditions significantly decreased the cellular proliferation, migration, invasion and angiogenesis of HUVECs. It was subsequently identified that TBX­3 mediated its effects through the activation of AKT and vascular endothelial growth factor (VEGF) signaling. However, the genetic knockdown of SIRT1 in the presence of TBX­3 overexpression and glucose failed to activate the AKT and VEGF signaling pathways. In conclusion, the results of the present study suggested that SIRT1 may positively regulate TBX­3 in endothelial cells, therefore, SIRT1 and/or TBX­3 may serve as potential novel biomarkers for disease progression.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Sirtuina 1/fisiología , Proteínas de Dominio T Box/fisiología , Adulto , Biomarcadores/metabolismo , Células Cultivadas , Femenino , Técnicas de Silenciamiento del Gen , Glucosa , Células Endoteliales de la Vena Umbilical Humana , Humanos , Masculino , Persona de Mediana Edad , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
J Dent Res ; 99(10): 1182-1191, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32442036

RESUMEN

The synchondrosis in the cranial base is an important growth center for the craniofacial region. Abnormalities in the synchondroses affect the development of adjacent regions, including the craniofacial skeleton. Here, we report that the transcription factor TBX1, the candidate gene for DiGeorge syndrome, is expressed in mesoderm-derived chondrocytes and plays an essential and specific role in spheno-occipital synchondrosis development by inhibiting the expression of genes involved in chondrocyte hypertrophy and osteogenesis. In Tbx1-deficient mice, the spheno-occipital synchondrosis was completely mineralized at birth. TBX1 interacts with RUNX2, a master molecule of osteoblastogenesis and a regulator of chondrocyte maturation, and suppresses its transcriptional activity. Indeed, deleting Tbx1 triggers accelerated mineralization due to accelerated chondrocyte differentiation, which is associated with ectopic expression of downstream targets of RUNX2 in the spheno-occipital synchondrosis. These findings reveal that TBX1 acts as a regulator of chondrocyte maturation and osteogenesis during the spheno-occipital synchondrosis development. Thus, the tight regulation of endochondral ossification by TBX1 is crucial for the normal progression of chondrocyte differentiation in the spheno-occipital synchondrosis.


Asunto(s)
Condrocitos , Condrogénesis , Hueso Occipital , Osteogénesis , Proteínas de Dominio T Box , Animales , Diferenciación Celular , Ratones , Hueso Esfenoides , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología
13.
Elife ; 92020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32238264

RESUMEN

The hippocampal dentate gyrus (DG) is a unique brain region maintaining neural stem cells (NCSs) and neurogenesis into adulthood. We used multiphoton imaging to visualize genetically defined progenitor subpopulations in live slices across key stages of mouse DG development, testing decades old static models of DG formation with molecular identification, genetic-lineage tracing, and mutant analyses. We found novel progenitor migrations, timings, dynamic cell-cell interactions, signaling activities, and routes underlie mosaic DG formation. Intermediate progenitors (IPs, Tbr2+) pioneered migrations, supporting and guiding later emigrating NSCs (Sox9+) through multiple transient zones prior to converging at the nascent outer adult niche in a dynamic settling process, generating all prenatal and postnatal granule neurons in defined spatiotemporal order. IPs (Dll1+) extensively targeted contacts to mitotic NSCs (Notch active), revealing a substrate for cell-cell contact support during migrations, a developmental feature maintained in adults. Mouse DG formation shares conserved features of human neocortical expansion.


Asunto(s)
Giro Dentado/embriología , Células-Madre Neurales/fisiología , Nicho de Células Madre/fisiología , Animales , Comunicación Celular , Movimiento Celular , Giro Dentado/citología , Ratones , Ratones Endogámicos C57BL , Neurogénesis/fisiología , Receptores Notch/fisiología , Transducción de Señal/fisiología , Proteínas de Dominio T Box/fisiología
14.
Mol Metab ; 36: 100965, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32240964

RESUMEN

OBJECTIVE: T-box 1 (TBX1) has been identified as a genetic marker of beige adipose tissue. TBX1 is a mesodermal development transcription factor essential for tissue patterning and cell fate determination. However, whether it plays a role in the process of adipose beiging or how it functions in adipose tissue has not been reported. Here, we examined the function of TBX1 in adipose tissue as well as adipose-derived stem cells from mice and humans. METHODS: Adipose-specific TBX1 transgenic (TBX1 AdipoTG) and adipose-specific TBX1 knockout (TBX1 AdipoKO) mice were generated to explore the function of TBX1 in the process of adipose beiging, metabolism and energy homeostasis in vivo. In vitro, we utilized a siRNA mediated approach to determine the function of TBX1 during adipogenesis in mouse and human stem cells. RESULTS: Adipose-specific overexpression of TBX1 was not sufficient to fully induce beiging and prevent diet-induced obesity. However, adipose TBX1 expression was necessary to defend body temperature during cold through regulation of UCP1 and for maintaining ß3-adrenergic sensitivity and glucose homeostasis in vivo. Loss of adipose TBX1 expression enhanced basal lipolysis and reduced the size of subcutaneous iWAT adipocytes. Reduction of TBX1 expression via siRNA significantly impaired adipogenesis of mouse stromal vascular cells but significantly enhanced adipogenesis in human adipose derived stem cells. CONCLUSIONS: Adipose expression of TBX1 is necessary, but not sufficient, to defend body temperature during cold via proper UCP1 expression. Adipose TBX1 expression was also required for proper insulin signaling in subcutaneous adipose as well as for maintaining ß-adrenergic sensitivity, but overexpression of TBX1 was not sufficient to induce adipocyte beiging or to prevent diet-induced obesity. TBX1 expression is enriched in adipose stem cells in which it has contrasting effects on adipogenesis in mouse versus human cells. Collectively, these data demonstrate the importance of adipose TBX1 in the regulation of beige adipocyte function, energy homeostasis, and adipocyte development.


Asunto(s)
Tejido Adiposo Beige/metabolismo , Grasa Subcutánea/metabolismo , Proteínas de Dominio T Box/metabolismo , Adipocitos Beige/metabolismo , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis/fisiología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Diferenciación Celular/genética , Regulación de la Expresión Génica/genética , Humanos , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Obesidad/metabolismo , Receptores Adrenérgicos beta/metabolismo , Transducción de Señal/genética , Grasa Subcutánea/fisiología , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Termogénesis/fisiología , Proteína Desacopladora 1/metabolismo
15.
Cell Rep ; 31(2): 107495, 2020 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-32294447

RESUMEN

Tbr1 is a high-confidence autism spectrum disorder (ASD) gene encoding a transcription factor with distinct pre- and postnatal functions. Postnatally, Tbr1 conditional knockout (CKO) mutants and constitutive heterozygotes have immature dendritic spines and reduced synaptic density. Tbr1 regulates expression of several genes that underlie synaptic defects, including a kinesin (Kif1a) and a WNT-signaling ligand (Wnt7b). Furthermore, Tbr1 mutant corticothalamic neurons have reduced thalamic axonal arborization. LiCl and a GSK3ß inhibitor, two WNT-signaling agonists, robustly rescue the dendritic spines and the synaptic and axonal defects, suggesting that this could have relevance for therapeutic approaches in some forms of ASD.


Asunto(s)
Espinas Dendríticas/metabolismo , Proteínas de Dominio T Box/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Trastorno del Espectro Autista/genética , Proteínas de Unión al ADN/metabolismo , Espinas Dendríticas/fisiología , Femenino , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Neurogénesis/fisiología , Neuronas/metabolismo , Neuronas/fisiología , Sinapsis/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/fisiología , Tálamo/metabolismo , Vía de Señalización Wnt/genética
16.
Dev Biol ; 461(2): 124-131, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32035085

RESUMEN

Development of multi-chambered heart is associated with spatio-temporal regulation of gene expression. A basic helix-loop-helix transcription factor Hey2 is specifically expressed in the embryonic mouse ventricles and is indispensable for ventricular myocyte differentiation, compartment identity and morphogenesis of the heart. However, how Hey2 transcription is precisely regulated in the heart remains unclear. In this study, we identified a distal Hey2 enhancer conserved in the mouse and human to possess specific transcriptional activity in ventricular free wall myocytes at the looping stage of cardiac development. Deletion of the enhancer significantly decreased endogenous Hey2 expression in the ventricular myocardium but not in other tissues of mouse embryos. Mutation/deletion of the conserved binding sites for T-box and Gata proteins, but not NK-2 proteins, abolished the enhancer activity, and Tbx20 null mice completely lost the enhancer activity in the embryonic ventricles. Luciferase reporter analysis suggested that the ventricular enhancer activity was controlled by Tbx20 through its DNA binding and cooperative function with cardiac Gata proteins. These results delineate a regulatory mechanism of ventricular Hey2 expression and help fully understand molecular cascades in myocardial cell differentiation and cardiac morphogenesis during embryonic development.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/biosíntesis , Elementos de Facilitación Genéticos , Factor de Transcripción GATA4/fisiología , Regulación del Desarrollo de la Expresión Génica , Ventrículos Cardíacos/embriología , Proteínas Represoras/biosíntesis , Proteínas de Dominio T Box/fisiología , Animales , Secuencia de Bases , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Secuencia Conservada , Genes Reporteros , Ventrículos Cardíacos/metabolismo , Humanos , Mamíferos/genética , Ratones , Ratones Transgénicos , Proteínas Represoras/genética , Alineación de Secuencia , Eliminación de Secuencia , Homología de Secuencia de Ácido Nucleico , Especificidad de la Especie
17.
Elife ; 92020 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-32039762

RESUMEN

RORγt+ group 3 innate lymphoid cells (ILC3s) maintain intestinal homeostasis through secretion of type 3 cytokines such as interleukin (IL)-17 and IL-22. However, CCR6- ILC3s additionally co-express T-bet allowing for the acquisition of type 1 effector functions. While T-bet controls the type 1 programming of ILC3s, the molecular mechanisms governing T-bet are undefined. Here, we identify c-Maf as a crucial negative regulator of murine T-bet+ CCR6- ILC3s. Phenotypic and transcriptomic profiling of c-Maf-deficient CCR6- ILC3s revealed a hyper type 1 differentiation status, characterized by overexpression of ILC1/NK cell-related genes and downregulation of type 3 signature genes. On the molecular level, c-Maf directly restrained T-bet expression. Conversely, c-Maf expression was dependent on T-bet and regulated by IL-1ß, IL-18 and Notch signals. Thus, we define c-Maf as a crucial cell-intrinsic brake in the type 1 effector acquisition which forms a negative feedback loop with T-bet to preserve the identity of CCR6- ILC3s.


Asunto(s)
Reprogramación Celular/fisiología , Inmunidad Innata , Linfocitos/metabolismo , Proteínas Proto-Oncogénicas c-maf/fisiología , Receptores CCR6/metabolismo , Proteínas de Dominio T Box/fisiología , Animales , Linaje de la Célula , Interleucina-18/fisiología , Interleucina-1beta/fisiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Regiones Promotoras Genéticas , Receptores Notch/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética
18.
Life Sci ; 239: 117040, 2019 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-31704448

RESUMEN

Congenital heart disease (CHD) is the most common birth defect in newborns. There is increasing evidence that apoptosis and remodeling of the cardiomyoblasts are the major pathology of CHD. Previous research found that T-box transcription factor 3 (TBX3) was compulsory for the regulation of proliferation, cell cycle arrest and apoptosis in various cells. Hence, TBX3 might be involved in the treatment of CHD. The primary aim of this study was to study the effects of TBX3 on apoptosis in aged cardiomyoblasts and investigate the latent mechanism. In the present study, we found TBX3 knockdown induced proliferation inhibition, cell cycle arrest and apoptosis accompanied by mitochondrial dysfunction in cardiomyoblasts at passage 10 to 15. Apoptosis-inducing effects of TBX3 silence could be neutralized by silencing P21 using specific siRNA. In addition, the mRNA and protein expression levels of TBX3 in the heart tissues of sporadic type CHD donors were obviously down-regulated. In conclusion, we demonstrated that TBX3 deficiency accelerated apoptosis via directly regulating P21 expression in senescent cardiomyoblasts.


Asunto(s)
Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/metabolismo , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular , Proliferación Celular/efectos de los fármacos , China , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Feto , Regulación de la Expresión Génica/genética , Cardiopatías Congénitas/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Miocitos Cardíacos/fisiología , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Ratas , Proteínas de Dominio T Box/fisiología , Factores de Transcripción/metabolismo
19.
Cell Rep ; 28(7): 1758-1772.e4, 2019 08 13.
Artículo en Inglés | MEDLINE | ID: mdl-31412245

RESUMEN

Following infection, inflammatory cues upregulate core transcriptional programs to establish pathogen-specific protection. In viral infections, T follicular helper (TFH) cells express the prototypical T helper 1 transcription factor T-bet. Several studies have demonstrated essential but conflicting roles for T-bet in TFH biology. Understanding the basis of this controversy is crucial, as modulation of T-bet expression instructs TFH differentiation and ultimately protective antibody responses. Comparing influenza and LCMV viral infections, we demonstrate that the role of T-bet is contingent on the environmental setting of TFH differentiation, IL-2 signaling, and T cell competition. Furthermore, we demonstrate that T-bet expression by either TFH or GC B cells independently drives antibody isotype class switching. Specifically, T cell-specific loss of T-bet promotes IgG1, whereas B cell-specific loss of T-bet inhibits IgG2a/c switching. Combined, this work highlights that the context-dependent induction of T-bet instructs the development of protective, neutralizing antibodies following viral infection or vaccination.


Asunto(s)
Formación de Anticuerpos/inmunología , Diferenciación Celular , Centro Germinal/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Orthomyxoviridae/inmunología , Proteínas de Dominio T Box/fisiología , Linfocitos T Colaboradores-Inductores/citología , Animales , Anticuerpos Antivirales/inmunología , Infecciones por Arenaviridae/inmunología , Infecciones por Arenaviridae/metabolismo , Infecciones por Arenaviridae/virología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Linfocitos B/virología , Femenino , Centro Germinal/metabolismo , Centro Germinal/virología , Inmunoglobulina G/metabolismo , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Coriomeningitis Linfocítica/virología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Transducción de Señal , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/virología
20.
Neuron ; 103(4): 642-657.e7, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31255486

RESUMEN

Neuronal subtypes show diverse injury responses, but the molecular underpinnings remain elusive. Using transgenic mice that allow reliable visualization of axonal fate, we demonstrate that intrinsically photosensitive retinal ganglion cells (ipRGCs) are both resilient to cell death and highly regenerative. Using RNA sequencing (RNA-seq), we show genes that are differentially expressed in ipRGCs and that associate with their survival and axon regeneration. Strikingly, thrombospondin-1 (Thbs1) ranked as the most differentially expressed gene, along with the well-documented injury-response genes Atf3 and Jun. THBS1 knockdown in RGCs eliminated axon regeneration. Conversely, RGC overexpression of THBS1 enhanced regeneration in both ipRGCs and non-ipRGCs, an effect that was dependent on syndecan-1, a known THBS1-binding protein. All structural domains of the THBS1 were not equally effective; the trimerization and C-terminal domains promoted regeneration, while the THBS type-1 repeats were dispensable. Our results identify cell-type-specific induction of Thbs1 as a novel gene conferring high regenerative capacity.


Asunto(s)
Regeneración Nerviosa/fisiología , Células Ganglionares de la Retina/fisiología , Trombospondina 1/fisiología , Animales , Apoptosis , Axones/metabolismo , Línea Celular , Femenino , Perfilación de la Expresión Génica , Genes Reporteros , Factor I del Crecimiento Similar a la Insulina/deficiencia , Factor I del Crecimiento Similar a la Insulina/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Compresión Nerviosa , Traumatismos del Nervio Óptico/genética , Traumatismos del Nervio Óptico/fisiopatología , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Opsinas de Bastones/deficiencia , Opsinas de Bastones/fisiología , Proteínas de Dominio T Box/deficiencia , Proteínas de Dominio T Box/fisiología , Trombospondina 1/biosíntesis , Trombospondina 1/genética , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA