Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 598
Filtrar
1.
Immunity ; 50(4): 924-940, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30995507

RESUMEN

Transforming growth factor (TGF)-ß is a crucial enforcer of immune homeostasis and tolerance, inhibiting the expansion and function of many components of the immune system. Perturbations in TGF-ß signaling underlie inflammatory diseases and promote tumor emergence. TGF-ß is also central to immune suppression within the tumor microenvironment, and recent studies have revealed roles in tumor immune evasion and poor responses to cancer immunotherapy. Here, we present an overview of the complex biology of the TGF-ß family and its context-dependent nature. Then, focusing on cancer, we discuss the roles of TGF-ß signaling in distinct immune cell types and how this knowledge is being leveraged to unleash the immune system against the tumor.


Asunto(s)
Neoplasias/inmunología , Factor de Crecimiento Transformador beta/fisiología , Inmunidad Adaptativa , Animales , Células Dendríticas/inmunología , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal , Fibroblastos/inmunología , Humanos , Inmunidad Innata , Inflamación , Macrófagos/inmunología , Ratones Noqueados , Neutrófilos/inmunología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/inmunología , Subgrupos de Linfocitos T/inmunología , Factor de Crecimiento Transformador beta/inmunología , Escape del Tumor , Microambiente Tumoral
2.
Immunity ; 47(5): 903-912.e4, 2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29126797

RESUMEN

Alveolar macrophages (AMs) derive from fetal liver monocytes, which colonize the lung during embryonic development and give rise to fully mature AMs perinatally. AM differentiation requires granulocyte macrophage colony-stimulating factor (GM-CSF), but whether additional factors are involved in AM regulation is not known. Here we report that AMs, in contrast to most other tissue macrophages, were also dependent on transforming growth factor-ß receptor (TGF-ßR) signaling. Conditional deletion of TGF-ßR in mice at different time points halted the development and differentiation of AMs. In adult mice, TGF-ß was also critical for AM homeostasis. The source of TGF-ß was AMs themselves, indicative of an autocrine loop that promotes AM self-maintenance. Mechanistically, TGF-ßR signaling resulted in upregulation of PPAR-γ, a signature transcription factor essential for the development of AMs. These findings reveal an additional layer of complexity regarding the guidance cues, which govern the genesis, maturation, and survival of AMs.


Asunto(s)
Homeostasis , Macrófagos Alveolares/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Diferenciación Celular , Desarrollo Embrionario , Ratones , Ratones Endogámicos C57BL , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/fisiología , Transcriptoma
3.
PLoS Genet ; 14(8): e1007496, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30133436

RESUMEN

During embryonic development, a number of genetic cues act to generate neuronal diversity. While intrinsic transcriptional cascades are well-known to control neuronal sub-type cell fate, the target cells can also provide critical input to specific neuronal cell fates. Such signals, denoted retrograde signals, are known to provide critical survival cues for neurons, but have also been found to trigger terminal differentiation of neurons. One salient example of such target-derived instructive signals pertains to the specification of the Drosophila FMRFamide neuropeptide neurons, the Tv4 neurons of the ventral nerve cord. Tv4 neurons receive a BMP signal from their target cells, which acts as the final trigger to activate the FMRFa gene. A recent FMRFa-eGFP genetic screen identified several genes involved in Tv4 specification, two of which encode components of the U5 subunit of the spliceosome: brr2 (l(3)72Ab) and Prp8. In this study, we focus on the role of RNA processing during target-derived signaling. We found that brr2 and Prp8 play crucial roles in controlling the expression of the FMRFa neuropeptide specifically in six neurons of the VNC (Tv4 neurons). Detailed analysis of brr2 revealed that this control is executed by two independent mechanisms, both of which are required for the activation of the BMP retrograde signaling pathway in Tv4 neurons: (1) Proper axonal pathfinding to the target tissue in order to receive the BMP ligand. (2) Proper RNA splicing of two genes in the BMP pathway: the thickveins (tkv) gene, encoding a BMP receptor subunit, and the Medea gene, encoding a co-Smad. These results reveal involvement of specific RNA processing in diversifying neuronal identity within the central nervous system.


Asunto(s)
Empalme Alternativo , Proteínas de Drosophila/fisiología , Drosophila/genética , FMRFamida/fisiología , Neuronas/fisiología , ARN Helicasas/fisiología , Factores de Empalme de ARN/fisiología , Animales , Diferenciación Celular , Sistema Nervioso Central/fisiología , Drosophila/fisiología , Proteínas de Drosophila/genética , FMRFamida/genética , Regulación del Desarrollo de la Expresión Génica , Mutación , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , ARN Helicasas/genética , Factores de Empalme de ARN/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/fisiología , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Análisis de Secuencia de ARN , Transducción de Señal , Empalmosomas , Factores de Transcripción/genética , Factores de Transcripción/fisiología
4.
Immunity ; 33(3): 313-25, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20870174

RESUMEN

The molecular mechanisms underlying retinoic acid (RA) augmentation of T cell receptor (TCR) and transforming growth factor-ß (TGF-ß)-induced Foxp3 transcription and inhibition of the latter by cytokines such as IL-27 were here shown to be related processes involving modifications of baseline (TGF-ß-induced) phosphorylated Smad3 (pSmad3) binding to a conserved enhancer region (enhancer I). RA augmentation involved the binding of retinoic acid receptor (RAR) and retinoid X receptor (RXR) to a dominant site in enhancer I and a subordinate site in the promoter. This led to increased histone acetylation in the region of the Smad3 binding site and increased binding of pSmad3. Cytokine (IL-27) inhibition involved binding of pStat3 to a gene silencer in a second conserved enhancer region (enhancer II) downstream from enhancer I; this led to loss of pSmad3 binding to enhancer I. Thus, control of accessibility and binding of pSmad3 provides a common framework for positive and negative regulation of TGF-ß-induced Foxp3 transcription.


Asunto(s)
Elementos de Facilitación Genéticos , Factores de Transcripción Forkhead/genética , Regulación de la Expresión Génica , Proteína smad3/metabolismo , Animales , Sitios de Unión , Femenino , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción NFATC/metabolismo , Fosforilación , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Antígenos de Linfocitos T/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Receptores X Retinoide/fisiología , Factor de Transcripción AP-1/fisiología , Transcripción Genética , Factor de Crecimiento Transformador beta/fisiología , Tretinoina/farmacología
5.
Proc Natl Acad Sci U S A ; 113(8): 2294-9, 2016 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-26842836

RESUMEN

Rupture of the ovarian follicle releases the oocyte at ovulation, a timed event that is critical for fertilization. It is not understood how the protease activity required for rupture is directed with precise timing and localization to the outer surface, or apex, of the follicle. We hypothesized that vasoconstriction at the apex is essential for rupture. The diameter and blood flow of individual vessels and the thickness of the apical follicle wall were examined over time to expected ovulation using intravital multiphoton microscopy. Vasoconstriction of apical vessels occurred within hours preceding follicle rupture in wild-type mice, but vasoconstriction and rupture were absent in Amhr2(cre/+)SmoM2 mice in which follicle vessels lack the normal association with vascular smooth muscle. Vasoconstriction is not simply a response to reduced thickness of the follicle wall; vasoconstriction persisted in wild-type mice when thinning of the follicle wall was prevented by infusion of protease inhibitors into the ovarian bursa. Ovulation was inhibited by preventing the periovulatory rise in the expression of the vasoconstrictor endothelin 2 by follicle cells of wild-type mice. In these mice, infusion of vasoconstrictors (either endothelin 2 or angiotensin 2) into the bursa restored the vasoconstriction of apical vessels and ovulation. Additionally, infusion of endothelin receptor antagonists into the bursa of wild-type mice prevented vasoconstriction and follicle rupture. Processing tissue to allow imaging at increased depth through the follicle and transabdominal ultrasonography in vivo showed that decreased blood flow is restricted to the apex. These results demonstrate that vasoconstriction at the apex of the follicle is essential for ovulation.


Asunto(s)
Folículo Ovárico/irrigación sanguínea , Folículo Ovárico/fisiología , Ovulación/fisiología , Vasoconstricción/fisiología , Animales , Endotelina-2/deficiencia , Endotelina-2/genética , Endotelina-2/fisiología , Femenino , Microscopía Intravital , Ratones , Ratones de la Cepa 129 , Ratones Transgénicos , Microscopía de Fluorescencia por Excitación Multifotónica , Folículo Ovárico/diagnóstico por imagen , Ovulación/genética , Receptores Acoplados a Proteínas G/deficiencia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiología , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Receptor Smoothened , Ultrasonografía , Vasoconstricción/genética
6.
Int J Cancer ; 142(5): 999-1009, 2018 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-29047105

RESUMEN

Immune evasion is a hallmark feature of cancer, and it plays an important role in tumour initiation and progression. In addition, tumour immune evasion severely hampers the desired antitumour effect in multiple cancers. In this study, we aimed to investigate the role of the Notch pathway in immune evasion in the head and neck squamous cell carcinoma (HNSCC) microenvironment. We first demonstrated that Notch1 signaling was activated in a Tgfbr1/Pten-knockout HNSCC mouse model. Notch signaling inhibition using a γ-secretase inhibitor (GSI-IX, DAPT) decreased tumour burden in the mouse model after prophylactic treatment. In addition, flow cytometry analysis indicated that Notch signaling inhibition reduced the sub-population of myeloid-derived suppressor cells (MDSCs), tumour-associated macrophages (TAMs) and regulatory T cells (Tregs), as well as immune checkpoint molecules (PD1, CTLA4, TIM3 and LAG3), in the circulation and in the tumour. Immunohistochemistry (IHC) of human HNSCC tissues demonstrated that elevation of the Notch1 downstream target HES1 was correlated with MDSC, TAM and Treg markers and with immune checkpoint molecules. These results suggest that modulating the Notch signaling pathway may decrease MDSCs, TAMs, Tregs and immune checkpoint molecules in HNSCC.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Carcinoma de Células Escamosas/inmunología , Diaminas/farmacología , Modelos Animales de Enfermedad , Neoplasias de Cabeza y Cuello/inmunología , Células Mieloides/inmunología , Linfocitos T Reguladores/inmunología , Tiazoles/farmacología , Animales , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de Cabeza y Cuello/metabolismo , Neoplasias de Cabeza y Cuello/patología , Humanos , Terapia de Inmunosupresión , Ratones , Ratones Noqueados , Células Mieloides/efectos de los fármacos , Fosfohidrolasa PTEN/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Notch1/genética , Receptor Notch1/metabolismo , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Linfocitos T Reguladores/efectos de los fármacos , Células Tumorales Cultivadas , Escape del Tumor/efectos de los fármacos
7.
Dev Biol ; 415(1): 14-23, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27180663

RESUMEN

The secondary palate separates the oral from the nasal cavity and its closure during embryonic development is sensitive to genetic perturbations. Mice with deleted Foxf2, encoding a forkhead transcription factor, are born with cleft palate, and an abnormal tongue morphology has been proposed as the underlying cause. Here, we show that Foxf2(-/-) maxillary explants cultured in vitro, in the absence of tongue and mandible, failed to close the secondary palate. Proliferation and collagen content were decreased in Foxf2(-/-) palatal shelf mesenchyme. Phosphorylation of Smad2/3 was reduced in mutant palatal shelf, diagnostic of attenuated canonical Tgfß signaling, whereas phosphorylation of p38 was increased. The amount of Tgfß2 protein was diminished, whereas the Tgfb2 mRNA level was unaltered. Expression of several genes encoding extracellular proteins important for Tgfß signaling were reduced in Foxf2(-)(/)(-) palatal shelves: a fibronectin splice-isoform essential for formation of extracellular Tgfß latency complexes; Tgfbr3 - or betaglycan - which acts as a co-receptor and an extracellular reservoir of Tgfß; and integrins αV and ß1, which are both Tgfß targets and required for activation of latent Tgfß. Decreased proliferation and reduced extracellular matrix content are consistent with diminished Tgfß signaling. We therefore propose that gene expression changes in palatal shelf mesenchyme that lead to reduced Tgfß signaling contribute to cleft palate in Foxf2(-)(/)(-) mice.


Asunto(s)
Fisura del Paladar/embriología , Factores de Transcripción Forkhead/fisiología , Mesodermo/embriología , Hueso Paladar/embriología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta2/fisiología , Animales , Colágeno/fisiología , Matriz Extracelular/fisiología , Proteínas de la Matriz Extracelular/fisiología , Fibronectinas/fisiología , Factores de Transcripción Forkhead/deficiencia , Factores de Transcripción Forkhead/genética , Regulación del Desarrollo de la Expresión Génica , Integrinas/fisiología , Mandíbula/embriología , Maxilar/embriología , Mesodermo/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Técnicas de Cultivo de Órganos , Fosforilación , Procesamiento Proteico-Postraduccional , Proteoglicanos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteína Smad2/fisiología , Proteína smad3/fisiología , Lengua/anomalías , Lengua/embriología , Factor de Crecimiento Transformador beta2/biosíntesis , Factor de Crecimiento Transformador beta2/genética
8.
Reproduction ; 153(4): 481-492, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28123059

RESUMEN

The influence of the hedgehog signaling pathway on reproduction was studied in transgenic mice in which a dominant active allele of the hedgehog signal transducer, smoothened (Smo), was conditionally expressed in the developing Müllerian duct and gonads through recombination mediated by anti-Müllerian hormone receptor 2-cre (Amhr2cre ). Previous studies showed that development of the oviduct and uterus are abnormal in female Amhr2cre/+SmoM2 mice. In the current study, focusing on mutant males, litter size was reduced 53% in crosses with wild-type females. An extra band of undifferentiated tissue extended along each epididymis and vas deferens, a position suggesting derivation from Müllerian ducts that failed to regress fully. Hedgehog signaling was elevated in this tissue, based on mRNA levels of target genes. Amhr2 mRNA was dramatically reduced in the uterus of mutant females and in the extra tissue in the tract of mutant males, suggesting that AMHR2 signaling was inadequate for complete Müllerian duct regression. Spermatogenesis and sperm motility were normal, but testis weight was reduced 37% and epididymal sperm number was reduced 36%. The number of sperm recovered from the uteri of wild-type females after mating with mutant males was reduced 78%. This suggested that sperm transport through the male tract was reduced, resulting in fewer sperm in the ejaculate. Consistent with this, mutant males had unusually tortuous vas deferentia with constrictions within the lumen. We concluded that persistence of a relatively undifferentiated remnant of Müllerian tissue is sufficient to cause subtle changes in the male reproductive tract that reduce fertility.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Proteínas Hedgehog/metabolismo , Infertilidad/patología , Conductos Paramesonéfricos/metabolismo , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Receptor Smoothened/fisiología , Animales , Epidídimo/citología , Epidídimo/metabolismo , Femenino , Infertilidad/etiología , Infertilidad/metabolismo , Integrasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Conductos Paramesonéfricos/citología , Reproducción/fisiología , Túbulos Seminíferos/citología , Túbulos Seminíferos/metabolismo , Transducción de Señal , Espermatogénesis
9.
J Am Soc Nephrol ; 27(10): 3006-3021, 2016 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26940097

RESUMEN

Organ-specific microRNAs have essential roles in maintaining normal organ function. However, the microRNA profile of the kidney and the role of microRNAs in modulating renal function remain undefined. We performed an unbiased assessment of the genome-wide microRNA expression profile in 14 mouse organs using Solexa deep sequencing and found that microRNA-196a (miR-196a) and miR-196b are selectively expressed in kidney, with 74.37% of mouse total miR-196a and 73.19% of mouse total miR-196b distributed in the kidneys. We confirmed the predominant expression of miR-196a/b in mouse and human kidney, particularly in the glomeruli and tubular epithelium, by quantitative RT-PCR and in situ hybridization assays. During unilateral ureteral obstruction (UUO)-induced mouse renal fibrosis, renal miR-196a/b levels rapidly decreased. Elevation of renal miR-196a/b expression by hydrodynamic-based delivery of a miR-196a/b-expressing plasmid before or shortly after UUO significantly downregulated profibrotic proteins, including collagen 1 and α-smooth muscle actin, and mitigated UUO-induced renal fibrosis. In contrast, depletion of renal miR-196a/b by miR-196a/b antagomirs substantially aggravated UUO-induced renal fibrosis. Mechanistic studies further identified transforming growth factor beta receptor II (TGFßR2) as a common target of miR-196a and miR-196b. Decreasing miR-196a/b expression in human HK2 cells strongly activated TGF-ß-Smad signaling and cell fibrosis; whereas increasing miR-196a/b levels in mouse primary cultured tubular epithelial cells inhibited TGF-ß-Smad signaling. In the UUO model, miR-196a/b silenced TGF-ß-Smad signaling, decreased the expression of collagen 1 and α-smooth muscle actin, and attenuated renal fibrosis. Our findings suggest that elevating renal miR-196a/b levels may be a novel therapeutic strategy for treating renal fibrosis.


Asunto(s)
Riñón/patología , MicroARNs/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Animales , Femenino , Fibrosis/genética , Humanos , Masculino , Ratones , Persona de Mediana Edad , Obstrucción Ureteral/genética
10.
J Formos Med Assoc ; 116(5): 351-358, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-27720345

RESUMEN

BACKGROUND/PURPOSE: In order to clarify the role of transforming growth factor beta 1 (TGF-ß1) in pulp repair/regeneration responses, we investigated the differential signaling pathways responsible for the effects of TGF-ß1 on collagen turnover, matrix metalloproteinase-3 (MMP-3), and tissue inhibitor of metalloproteinase-1 (TIMP-1) production in human dental pulp cells. METHODS: Pulp cells were exposed to TGF-ß1 with/without pretreatment and coincubation by 1,4-diamino-2,3-dicyano-1,4-bis(o-aminophenyl mercapto)butadiene (U0126; a mitogen-activated protein kinase kinase [MEK]/extracellular signal-regulated kinase [ERK] inhibitor) and 4-(5-benzol[1,3]dioxol-5-yl-4-pyrldin-2-yl-1H- imidazol-2-yl)-benzamide hydrate (SB431542; an activin receptor-like kinase-5/Smad signaling inhibitor). Sircol collagen assay was used to measure cellular collagen content. Culture medium procollagen I, TIMP-1, and MMP-3 levels were determined by enzyme-linked immunosorbent assay. RESULTS: TGF-ß1 increased the collagen content, procollagen I, and TIMP-1 production, but slightly decreased MMP-3 production of pulp cells. SB431542 and U0126 prevented the TGF-ß1-induced increase of collagen content and TIMP-1 production of dental pulp cells. CONCLUSION: These results indicate that TGF-ß1 may be involved in the healing/regeneration processes of dental pulp in response to injury by stimulation of collagen and TIMP-1 production. These events are associated with activin receptor-like kinase-5/Smad2/3 and MEK/ERK signaling.


Asunto(s)
Colágeno Tipo I/metabolismo , Colágeno/fisiología , Regeneración/fisiología , Inhibidor Tisular de Metaloproteinasa-1/metabolismo , Factor de Crecimiento Transformador beta1/fisiología , Benzamidas/farmacología , Butadienos/farmacología , Células Cultivadas , Pulpa Dental/citología , Dioxoles/farmacología , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/fisiología , Nitrilos/farmacología , Proteínas Serina-Treonina Quinasas/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/efectos de los fármacos , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Regeneración/efectos de los fármacos , Proteínas Smad/efectos de los fármacos , Proteínas Smad/fisiología
11.
J Physiol ; 594(13): 3575-88, 2016 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-27006168

RESUMEN

KEY POINTS: The sensory components of the urinary bladder are responsible for the transduction of bladder filling and are often impaired with neurological injury or disease. Elevated extracellular ATP contributes, in part, to bladder afferent nerve hyperexcitability during urinary bladder inflammation or irritation. Transforming growth factor-ß1 (TGF-ß1) may stimulate ATP release from the urothelium through vesicular exocytosis mechanisms with minimal contribution from pannexin-1 channels to increase bladder afferent nerve discharge. Bladder afferent nerve hyperexcitability and urothelial ATP release with CYP-induced cystitis is decreased with TGF-ß inhibition. These results establish a causal link between an inflammatory mediator, TGF-ß, and intrinsic signalling mechanisms of the urothelium that may contribute to the altered sensory processing of bladder filling. ABSTRACT: The afferent limb of the micturition reflex is often compromised following bladder injury, disease and inflammatory conditions. We have previously demonstrated that transforming growth factor-ß (TGF-ß) signalling contributes to increased voiding frequency and decreased bladder capacity with cystitis. Despite the functional presence of TGF-ß in bladder inflammation, the precise mechanisms of TGF-ß mediating bladder dysfunction are not yet known. Thus, the present studies investigated the sensory components of the urinary bladder that may underlie the pathophysiology of aberrant TGF-ß activation. We utilized bladder-pelvic nerve preparations to characterize bladder afferent nerve discharge and the mechanisms of urothelial ATP release with distention. Our findings indicate that bladder afferent nerve discharge is sensitive to elevated extracellular ATP during pathological conditions of urinary bladder inflammation or irritation. We determined that TGF-ß1 may increase bladder afferent nerve excitability by stimulating ATP release from the urothelium via vesicular exocytosis mechanisms with minimal contribution from pannexin-1 channels. Furthermore, blocking aberrant TGF-ß signalling in cyclophosphamide-induced cystitis with TßR-1 inhibition decreased afferent nerve hyperexcitability with a concomitant decrease in urothelial ATP release. Taken together, these results establish a role for purinergic signalling mechanisms in TGF-ß-mediated bladder afferent nerve activation that may ultimately facilitate increased voiding frequency. The synergy between intrinsic urinary bladder signalling mechanisms and an inflammatory mediator provides novel insight into bladder dysfunction and supports new avenues for therapeutic intervention.


Asunto(s)
Adenosina Trifosfato/fisiología , Cistitis/fisiopatología , Receptores Purinérgicos/fisiología , Factor de Crecimiento Transformador beta/fisiología , Vejiga Urinaria/inervación , Vejiga Urinaria/fisiología , Animales , Conexinas/fisiología , Ciclofosfamida , Cistitis/inducido químicamente , Masculino , Ratones Endogámicos C57BL , Proteínas del Tejido Nervioso/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Antagonistas Purinérgicos/farmacología , Fosfato de Piridoxal/análogos & derivados , Fosfato de Piridoxal/farmacología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Urotelio/fisiología
12.
Exp Dermatol ; 25(2): 85-91, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26476131

RESUMEN

Keratoacanthoma (KA) are common but exceptional benign tumors, often appearing on sun-exposed areas of light skinned people and showing spontaneous resolution. The goal of this study was to review existing literature, to point out the etiological complexity of KA biology and to answer the controversial debate if or not KA is a distinct entity or a variant of squamous cell carcinoma (SCC). Relying on recent results, we highlight that KA is an individual lesion with a unique molecular signature caused by alterations in the TGFß signalling pathway. These recent findings will help to understand the nature of KA and to develop new reliable diagnostic tools, simplifying the discrimination of the histologically similar KA and SCC.


Asunto(s)
Queratoacantoma , Enfermedades de la Piel , Carcinoma de Células Escamosas/diagnóstico , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/efectos de la radiación , Hibridación Genómica Comparativa , Diagnóstico Diferencial , Progresión de la Enfermedad , Predisposición Genética a la Enfermedad , Humanos , Queratoacantoma/diagnóstico , Queratoacantoma/etiología , Queratoacantoma/genética , Queratoacantoma/metabolismo , Queratoacantoma/patología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/fisiología , Neoplasias Inducidas por Radiación/química , Neoplasias Inducidas por Radiación/diagnóstico , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal , Enfermedades de la Piel/diagnóstico , Enfermedades de la Piel/etiología , Enfermedades de la Piel/genética , Enfermedades de la Piel/metabolismo , Enfermedades de la Piel/patología , Neoplasias Cutáneas/diagnóstico , Luz Solar/efectos adversos , Factor de Crecimiento Transformador beta/fisiología , Rayos Ultravioleta/efectos adversos
13.
J Neurochem ; 132(4): 418-28, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25421849

RESUMEN

The mechanism by which extracellular molecules control serotonergic cell fate remains elusive. Recently, we showed that noggin, which inactivates bone morphogenetic proteins (BMPs), induces serotonergic differentiation of mouse embryonic (ES) and induced pluripotent stem cells with coordinated gene expression along the serotonergic lineage. Here, we created a rapid assay for serotonergic induction by generating knock-in ES cells expressing a naturally secreted Gaussia luciferase driven by the enhancer of Pet-1/Fev, a landmark of serotonergic differentiation. Using these cells, we performed candidate-based screening and identified BMP type I receptor kinase inhibitors LDN-193189 and DMH1 as activators of luciferase. LDN-193189 induced ES cells to express the genes encoding Pet-1, tryptophan hydroxylase 2, and the serotonin transporter, and increased serotonin release without altering dopamine release. In contrast, TGF-ß receptor inhibitor SB-431542 selectively inhibited serotonergic differentiation, without changing overall neuronal differentiation. LDN-193189 inhibited expression of the BMP signaling target gene Id, and induced the TGF-ß target gene Lefty, whereas the opposite effect was observed with SB-431542. This study thus provides a new tool to investigate serotonergic differentiation and suggests that inhibition of BMP type I receptors and concomitant activation of TGF-ß receptor signaling are implicated in serotonergic differentiation. Candidate-based screening for serotonergic induction using a rapid assay in mouse embryonic stem cells revealed that the bone morphogenetic protein (BMP) type I receptor kinase inhibitors selectively induce serotonergic differentiation, whereas the TGF-ß receptor inhibitor SB-431542 inhibits the differentiation. These results suggest that inhibition of BMP type I receptors and concomitant activation of transforming growth factor-ß (TGF-ß) receptor signaling are involved in the early trajectory of serotonergic differentiation.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/fisiología , Diferenciación Celular/fisiología , Células Madre Embrionarias/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Neuronas Serotoninérgicas/fisiología , Animales , Benzamidas/farmacología , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/antagonistas & inhibidores , Técnicas de Cultivo de Célula/métodos , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dioxoles/farmacología , Células Madre Embrionarias/efectos de los fármacos , Técnicas de Sustitución del Gen/métodos , Ratones , Pirazoles/farmacología , Pirimidinas/farmacología , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Neuronas Serotoninérgicas/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factores de Tiempo
14.
Immunol Cell Biol ; 93(7): 662-72, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25823994

RESUMEN

Although transforming growth factor-ß (TGF-ß) has been shown to positively regulate the development of murine T helper type 17 (Th17) cells, which of the intracellular signaling pathways are involved is controversial. We examined Smad-dependent and -independent signaling molecules downstream of the TGF-ß receptor (TGFßR) involved in Th17 differentiation of naive murine CD4(+)CD62L(+) T cells. During Th17 differentiation of wild-type T cells, Smad2/3 was phosphorylated, indicating activation of the canonical Smad pathway. T cells lacking TGFßRII did not differentiate into Th17, whereas T cells treated with a TGFßRI kinase inhibitor (SB-431542) or overexpression of inhibitory Smad7 retained a low amount of Th17 polarization despite absent Smad2/3 phosphorylation. Using protein antibody arrays we found an increase of expression and phosphorylation of the following Smad-independent signaling molecules in Th17-polarized wild-type T cells: AKT1(Tyr474), AKT2 (Ser474), ERK1-p44/42 MAPK(Tyr204), mTOR(Thr2446), p38 MAPK(Thr180), Rac1/cdc42(Ser71), SAPK/JNK(Tyr185) and SP1(Thr739). Pharmacological inhibition of AKT/mammalian target of rapamycin (mTOR) signaling with rapamycin or LY294002 decreased Th17 differentiation of wild-type T cells, and completely abolished interleukin-17 production in T cells with overexpression of Smad7. Rapamycin and LY294002 also decreased induced regulatory T cell differentiation, but only had minor additive effects to Smad7 overexpression. Finally, inhibitors of mitogen-activated protein kinase (MAPK) blocked in vitro polarization of Th17 cells. Our data show that Smad-dependent and -independent intracellular pathways contribute to murine Th17 differentiation.


Asunto(s)
Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/fisiología , Células Th17/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Cromonas/farmacología , Interleucina-17/biosíntesis , Interleucina-17/genética , Linfopoyesis/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Morfolinas/farmacología , Fosfatidilinositol 3-Quinasas/fisiología , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Proto-Oncogénicas c-akt/fisiología , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/antagonistas & inhibidores , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Proteínas Smad/fisiología , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Serina-Treonina Quinasas TOR/fisiología , Células Th17/citología
15.
Hepatology ; 59(2): 483-95, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23996730

RESUMEN

UNLABELLED: Transforming growth factor beta (TGF-ß) signaling activates Smad- and TGF-ß-activated kinase 1 (TAK1)-dependent signaling to regulate cell survival, proliferation, fibrosis, and tumorigenesis. The effects of TGF-ß signaling on metabolic syndrome, including nonalcoholic fatty liver disease, remain elusive. Wild-type (WT) and hepatocyte-specific TGF-ß receptor type II-deficient (Tgfbr2ΔHEP) mice were fed a choline-deficient amino acid (CDAA)-defined diet for 22 weeks to induce NASH. WT mice fed a CDAA diet displayed increased activation of Smad2/3 and had marked lipid accumulation, inflammatory cell infiltration, hepatocyte death, and fibrosis; in comparison, Tgfbr2ΔHEP mice fed a CDAA diet had suppressed liver steatosis, inflammation, and fibrosis. Both palmitate-induced steatotic hepatocytes and hepatocytes isolated from WT mice fed a CDAA diet had increased susceptibility to TGF-ß-mediated death. TGF-ß-mediated death in steatotic hepatocytes was inhibited by silencing Smad2 or blocking reactive oxygen species (ROS) production and was enhanced by inhibiting TAK1 or nuclear factor kappa B. Increased hepatic steatosis in WT mice fed a CDAA diet was associated with the increased expression of lipogenesis genes (Dgat1 and Srebp1c), whereas the decreased steatosis in Tgfbr2ΔHEP mice was accompanied by the increased expression of genes involved in ß-oxidation (Cpt1 and Acox1). In combination with palmitate treatment, TGF-ß signaling promoted lipid accumulation with induction of lipogenesis-related genes and suppression of ß-oxidation-related genes in hepatocytes. Silencing Smad2 decreased TGF-ß-mediated lipid accumulation and corrected altered gene expression related to lipid metabolism in hepatocytes. Finally, we confirmed that livers from patients with nonalcoholic steatohepatitis (NASH) displayed phosphorylation and nuclear translocation of Smad2/3. CONCLUSIONS: TGF-ß signaling in hepatocytes contributes to hepatocyte death and lipid accumulation through Smad signaling and ROS production that promote the development of NASH.


Asunto(s)
Apoptosis/fisiología , Hígado Graso/fisiopatología , Hepatocitos/fisiología , Metabolismo de los Lípidos/fisiología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/fisiología , Animales , Células Cultivadas , Deficiencia de Colina/metabolismo , Deficiencia de Colina/fisiopatología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Hígado Graso/etiología , Hígado Graso/patología , Eliminación de Gen , Hepatocitos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/fisiología , Especies Reactivas de Oxígeno/metabolismo , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Proteínas Smad/fisiología
16.
Biochem J ; 460(1): 35-47, 2014 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-24597762

RESUMEN

GDF15 (growth/differentiation factor 15), a novel member of the TGFß (transforming growth factor ß) superfamily, plays critical roles in the central and peripheral nervous systems, but the signal transduction pathways and receptor subtypes involved are not well understood. In the present paper, we report that GDF15 specifically increases the IK (delayed-rectifier outward K+ current) in rat CGNs (cerebellar granule neurons) in time- and concentration-dependent manners. The GDF15-induced amplification of the IK is mediated by the increased expression and reduced lysosome-dependent degradation of the Kv2.1 protein, the main α-subunit of the IK channel. Exposure of CGNs to GDF15 markedly induced the phosphorylation of ERK (extracellular-signal-regulated kinase), Akt and mTOR (mammalian target of rapamycin), but the GDF15-induced IK densities and increased expression of Kv2.1 were attenuated only by Akt and mTOR, and not ERK, inhibitors. Pharmacological inhibition of the Src-mediated phosphorylation of TGFßR2 (TGFß receptor 2), not TGFßR1, abrogated the effect of GDF15 on IK amplification and Kv2.1 induction. Immunoprecipitation assays showed that GDF15 increased the tyrosine phosphorylation of TGFßRII in the CGN lysate. The results of the present study reveal a novel regulation of Kv2.1 by GDF15 mediated through the TGFßRII-activated Akt/mTOR pathway, which is a previously uncharacterized Smad-independent mechanism of GDF15 signalling.


Asunto(s)
Cerebelo/fisiología , Factor 15 de Diferenciación de Crecimiento/fisiología , Proteína Oncogénica v-akt/fisiología , Proteínas Serina-Treonina Quinasas/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Canales de Potasio Shab/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/fisiología , Animales , Animales Recién Nacidos , Células Cultivadas , Cerebelo/citología , Humanos , Ratas , Ratas Sprague-Dawley , Receptor Tipo II de Factor de Crecimiento Transformador beta , Transducción de Señal/genética
17.
PLoS Genet ; 8(7): e1002798, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22807687

RESUMEN

Heterogametic sex chromosomes have evolved independently in various lineages of vertebrates. Such sex chromosome pairs often contain nonrecombining regions, with one of the chromosomes harboring a master sex-determining (SD) gene. It is hypothesized that these sex chromosomes evolved from a pair of autosomes that diverged after acquiring the SD gene. By linkage and association mapping of the SD locus in fugu (Takifugu rubripes), we show that a SNP (C/G) in the anti-Müllerian hormone receptor type II (Amhr2) gene is the only polymorphism associated with phenotypic sex. This SNP changes an amino acid (His/Asp384) in the kinase domain. While females are homozygous (His/His384), males are heterozygous. Sex in fugu is most likely determined by a combination of the two alleles of Amhr2. Consistent with this model, the medaka hotei mutant carrying a substitution in the kinase domain of Amhr2 causes a female phenotype. The association of the Amhr2 SNP with phenotypic sex is conserved in two other species of Takifugu but not in Tetraodon. The fugu SD locus shows no sign of recombination suppression between X and Y chromosomes. Thus, fugu sex chromosomes represent an unusual example of proto-sex chromosomes. Such undifferentiated X-Y chromosomes may be more common in vertebrates than previously thought.


Asunto(s)
Sustitución de Aminoácidos/genética , Receptores de Péptidos/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Procesos de Determinación del Sexo/genética , Takifugu , Animales , Evolución Biológica , Femenino , Estudios de Asociación Genética , Ligamiento Genético , Heterocigoto , Homocigoto , Masculino , Mutación Missense/genética , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Cromosomas Sexuales/genética , Takifugu/genética , Takifugu/fisiología
18.
Development ; 138(10): 1967-75, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21490063

RESUMEN

During male sexual differentiation, the transforming growth factor-ß (TGF-ß) signaling molecule anti-Müllerian hormone (AMH; also known as Müllerian inhibiting substance, MIS) is secreted by the fetal testes and induces regression of the Müllerian ducts, the primordia of the female reproductive tract organs. Currently, the molecular identity of downstream events regulated by the AMH signaling pathway remains unclear. We found that male-specific Wnt4 expression in mouse Müllerian duct mesenchyme depends upon AMH signaling, implicating the WNT pathway as a downstream mediator of Müllerian duct regression. Inactivation of ß-catenin, a mediator of the canonical WNT pathway, did not affect AMH signaling activation in the Müllerian duct mesenchyme, but did block Müllerian duct regression. These data suggest that ß-catenin mediates AMH signaling for Müllerian duct regression during male sexual differentiation.


Asunto(s)
Conductos Paramesonéfricos/embriología , Diferenciación Sexual/fisiología , Testículo/embriología , beta Catenina/fisiología , Animales , Hormona Antimülleriana/fisiología , Secuencia de Bases , Cartilla de ADN/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genitales Femeninos/embriología , Genitales Femeninos/metabolismo , Masculino , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Modelos Biológicos , Conductos Paramesonéfricos/metabolismo , Embarazo , Receptores de Péptidos/deficiencia , Receptores de Péptidos/genética , Receptores de Péptidos/fisiología , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Diferenciación Sexual/genética , Transducción de Señal , Testículo/metabolismo , Proteínas Wnt/deficiencia , Proteínas Wnt/genética , Proteínas Wnt/fisiología , Proteína Wnt4 , beta Catenina/deficiencia , beta Catenina/genética
19.
Blood ; 119(15): 3486-94, 2012 Apr 12.
Artículo en Inglés | MEDLINE | ID: mdl-22371886

RESUMEN

Invariant natural killer T (iNKT) cells have the ability to rapidly secret cytokines in response to diverse stimuli, and therefore influence numerous immune reactions. Although IFN-γ and IL-4 are thought to dominate iNKT cytokine production, a distinct subset of iNKT cells, expressing RORγt and producing IL-17, has now been identified in both mice and humans. Although a role in pathogen and allergic responses has been assigned to the RORγt(+) iNKT subset, factors controlling their development and function remain illusive. Here, we demonstrate that RORγt(+) iNKT-cell differentiation obeys transforming growth factor-ß (TGF-ß) signaling control, different from that described for conventional iNKT cells. We reveal that TGF-ß signaling, and particularly its SMAD4-dependent pathway, is required for both the survival of RORγt(+) iNKT cells during their development and IL-17 production at the periphery. Moreover, constitutive TGF-ß signaling in RORγt(+) iNKT cells drives higher peripheral numbers and increased tissue distribution. Finally, we found that SMAD4-dependent TGF-ß signaling is mandatory for the peripheral expansion of the RORγt(+) iNKT cells responding to inflammatory signals. Thus, this work demonstrates that both the development and responsiveness of the newly described IL-17-producing iNKT cell subset is under the control of a dedicated TGF-ß signaling pathway.


Asunto(s)
Diferenciación Celular , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/fisiología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Factor de Crecimiento Transformador beta/fisiología , Animales , Apoptosis/genética , Apoptosis/fisiología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Proliferación Celular/efectos de los fármacos , Interleucina-17/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células T Asesinas Naturales/efectos de los fármacos , Células T Asesinas Naturales/metabolismo , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/genética , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Transducción de Señal/genética , Transducción de Señal/inmunología , Transducción de Señal/fisiología , Proteína Smad4/genética , Proteína Smad4/metabolismo , Timocitos/metabolismo , Timocitos/fisiología , Timo/citología , Timo/efectos de los fármacos , Timo/metabolismo , Timo/fisiología , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/farmacología
20.
J Immunol ; 189(8): 3878-93, 2012 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-22972928

RESUMEN

In vitro data and transgenic mouse models suggest a role for TGF-ß signaling in dendritic cells (DCs) to prevent autoimmunity primarily through maintenance of DCs in their immature and tolerogenic state characterized by low expression of MHC class II (MHCII) and costimulatory molecules and increased expression of IDO, among others. To test whether a complete lack of TGF-ß signaling in DCs predisposes mice to spontaneous autoimmunity and to verify the mechanisms implicated previously in vitro, we generated conditional knockout (KO) mice with Cre-mediated DC-specific deletion of Tgfbr2 (DC-Tgfbr2 KO). DC-Tgfbr2 KO mice die before 15 wk of age with multiorgan autoimmune inflammation and spontaneous activation of T and B cells. Interestingly, there were no significant differences in the expression of MHCII, costimulatory molecules, or IDO in secondary lymphoid organ DCs, although Tgfbr2-deficient DCs were more proinflammatory in vitro and in vivo. DC-Tgfbr2 KO showed attenuated Foxp3 expression in regulatory T cells (Tregs) and abnormal expansion of CD25(-)Foxp3(+) Tregs in vivo. Tgfbr2-deficient DCs secreted elevated levels of IFN-γ and were not capable of directing Ag-specific Treg conversion unless in the presence of anti-IFN-γ blocking Ab. Adoptive transfer of induced Tregs into DC-Tgfbr2 KO mice partially rescued the phenotype. Therefore, in vivo, TGF-ß signaling in DCs is critical in the control of autoimmunity through both Treg-dependent and -independent mechanisms, but it does not affect MHCII and costimulatory molecule expression.


Asunto(s)
Enfermedades Autoinmunes/prevención & control , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Proteínas Serina-Treonina Quinasas/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Colitis/genética , Colitis/inmunología , Colitis/prevención & control , Células Dendríticas/patología , Modelos Animales de Enfermedad , Tolerancia Inmunológica/genética , Inmunofenotipificación , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Especificidad de Órganos/genética , Especificidad de Órganos/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Receptor Tipo II de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/fisiología , Linfocitos T Reguladores/patología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda