Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24.075
Filtrar
Más filtros

Intervalo de año de publicación
1.
Annu Rev Immunol ; 34: 369-94, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27168242

RESUMEN

Vitamin A is a multifunctional vitamin implicated in a wide range of biological processes. Its control over the immune system and functions are perhaps the most pleiotropic not only for development but also for the functional fate of almost every cell involved in protective or regulatory adaptive or innate immunity. This is especially key at the intestinal border, where dietary vitamin A is first absorbed. Most effects of vitamin A are exerted by its metabolite, retinoic acid (RA), which through ligation of nuclear receptors controls transcriptional expression of RA target genes. In addition to this canonical function, RA and RA receptors (RARs), either as ligand-receptor or separately, play extranuclear, nongenomic roles that greatly expand the multiple mechanisms employed for their numerous and paradoxical functions that ultimately link environmental sensing with immune cell fate. This review discusses RA and RARs and their complex roles in innate and adaptive immunity.


Asunto(s)
Sistema Inmunológico , Mucosa Intestinal/fisiología , Receptores de Ácido Retinoico/inmunología , Tretinoina/metabolismo , Vitamina A/inmunología , Inmunidad Adaptativa , Animales , Humanos , Inmunidad Innata , Inmunomodulación , Receptores de Ácido Retinoico/metabolismo , Tretinoina/inmunología
2.
Cell ; 180(6): 1098-1114.e16, 2020 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-32169218

RESUMEN

The immunosuppressive tumor microenvironment (TME) is a major barrier to immunotherapy. Within solid tumors, why monocytes preferentially differentiate into immunosuppressive tumor-associated macrophages (TAMs) rather than immunostimulatory dendritic cells (DCs) remains unclear. Using multiple murine sarcoma models, we find that the TME induces tumor cells to produce retinoic acid (RA), which polarizes intratumoral monocyte differentiation toward TAMs and away from DCs via suppression of DC-promoting transcription factor Irf4. Genetic inhibition of RA production in tumor cells or pharmacologic inhibition of RA signaling within TME increases stimulatory monocyte-derived cells, enhances T cell-dependent anti-tumor immunity, and synergizes with immune checkpoint blockade. Furthermore, an RA-responsive gene signature in human monocytes correlates with an immunosuppressive TME in multiple human tumors. RA has been considered as an anti-cancer agent, whereas our work demonstrates its tumorigenic capability via myeloid-mediated immune suppression and provides proof of concept for targeting this pathway for tumor immunotherapy.


Asunto(s)
Monocitos/inmunología , Tretinoina/metabolismo , Microambiente Tumoral/inmunología , Animales , Carcinogénesis/patología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Línea Celular Tumoral , Células Dendríticas/inmunología , Humanos , Terapia de Inmunosupresión/métodos , Inmunoterapia/métodos , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Monocitos/metabolismo
3.
Cell ; 169(5): 807-823.e19, 2017 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-28479188

RESUMEN

Dormant hematopoietic stem cells (dHSCs) are atop the hematopoietic hierarchy. The molecular identity of dHSCs and the mechanisms regulating their maintenance or exit from dormancy remain uncertain. Here, we use single-cell RNA sequencing (RNA-seq) analysis to show that the transition from dormancy toward cell-cycle entry is a continuous developmental path associated with upregulation of biosynthetic processes rather than a stepwise progression. In addition, low Myc levels and high expression of a retinoic acid program are characteristic for dHSCs. To follow the behavior of dHSCs in situ, a Gprc5c-controlled reporter mouse was established. Treatment with all-trans retinoic acid antagonizes stress-induced activation of dHSCs by restricting protein translation and levels of reactive oxygen species (ROS) and Myc. Mice maintained on a vitamin A-free diet lose HSCs and show a disrupted re-entry into dormancy after exposure to inflammatory stress stimuli. Our results highlight the impact of dietary vitamin A on the regulation of cell-cycle-mediated stem cell plasticity. VIDEO ABSTRACT.


Asunto(s)
Células Madre Hematopoyéticas/citología , Transducción de Señal , Tretinoina/farmacología , Vitamina A/administración & dosificación , Animales , Vías Biosintéticas , Técnicas de Cultivo de Célula , Ciclo Celular/efectos de los fármacos , Supervivencia Celular , Dieta , Perfilación de la Expresión Génica , Células Madre Hematopoyéticas/efectos de los fármacos , Ratones , Poli I-C/farmacología , Especies Reactivas de Oxígeno/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Análisis de la Célula Individual , Estrés Fisiológico , Vitamina A/farmacología , Vitaminas/administración & dosificación , Vitaminas/farmacología
4.
Genes Dev ; 38(7-8): 308-321, 2024 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-38719541

RESUMEN

The transcription factor Oct4/Pou5f1 is a component of the regulatory circuitry governing pluripotency and is widely used to induce pluripotency from somatic cells. Here we used domain swapping and mutagenesis to study Oct4's reprogramming ability, identifying a redox-sensitive DNA binding domain, cysteine residue (Cys48), as a key determinant of reprogramming and differentiation. Oct4 Cys48 sensitizes the protein to oxidative inhibition of DNA binding activity and promotes oxidation-mediated protein ubiquitylation. Pou5f1 C48S point mutation has little effect on undifferentiated embryonic stem cells (ESCs) but upon retinoic acid (RA) treatment causes retention of Oct4 expression, deregulated gene expression, and aberrant differentiation. Pou5f1 C48S ESCs also form less differentiated teratomas and contribute poorly to adult somatic tissues. Finally, we describe Pou5f1 C48S (Janky) mice, which in the homozygous condition are severely developmentally restricted after E4.5. Rare animals bypassing this restriction appear normal at birth but are sterile. Collectively, these findings uncover a novel Oct4 redox mechanism involved in both entry into and exit from pluripotency.


Asunto(s)
Diferenciación Celular , Reprogramación Celular , Factor 3 de Transcripción de Unión a Octámeros , Oxidación-Reducción , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Factor 3 de Transcripción de Unión a Octámeros/genética , Animales , Ratones , Diferenciación Celular/genética , Reprogramación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Tretinoina/farmacología , Tretinoina/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Humanos
5.
Immunity ; 55(1): 129-144.e8, 2022 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-34910930

RESUMEN

Dendritic cells (DCs) patrol tissues and transport antigens to lymph nodes to initiate adaptive immune responses. Within tissues, DCs constitute a complex cell population composed of distinct subsets that can exhibit different activation states and functions. How tissue-specific cues orchestrate DC diversification remains elusive. Here, we show that the small intestine included two pools of cDC2s originating from common pre-DC precursors: (1) lamina propria (LP) CD103+CD11b+ cDC2s that were mature-like proinflammatory cells and (2) intraepithelial cDC2s that exhibited an immature-like phenotype as well as tolerogenic properties. These phenotypes resulted from the action of food-derived retinoic acid (ATRA), which enhanced actomyosin contractility and promoted LP cDC2 transmigration into the epithelium. There, cDC2s were imprinted by environmental cues, including ATRA itself and the mucus component Muc2. Hence, by reaching distinct subtissular niches, DCs can exist as immature and mature cells within the same tissue, revealing an additional mechanism of DC functional diversification.


Asunto(s)
Células Dendríticas/inmunología , Inflamación/inmunología , Mucosa Intestinal/patología , Linfocitos T/inmunología , Actomiosina/metabolismo , Animales , Presentación de Antígeno , Antígenos CD/metabolismo , Antígeno CD11b/metabolismo , Diferenciación Celular , Movimiento Celular , Células Cultivadas , Tolerancia Inmunológica , Cadenas alfa de Integrinas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucina 2/inmunología , Tretinoina/metabolismo
6.
Nat Immunol ; 19(12): 1403-1414, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30397350

RESUMEN

Repair of tissue damaged during inflammatory processes is key to the return of local homeostasis and restoration of epithelial integrity. Here we describe CD161+ regulatory T (Treg) cells as a distinct, highly suppressive population of Treg cells that mediate wound healing. These Treg cells were enriched in intestinal lamina propria, particularly in Crohn's disease. CD161+ Treg cells had an all-trans retinoic acid (ATRA)-regulated gene signature, and CD161 expression on Treg cells was induced by ATRA, which directly regulated the CD161 gene. CD161 was co-stimulatory, and ligation with the T cell antigen receptor induced cytokines that accelerated the wound healing of intestinal epithelial cells. We identified a transcription-factor network, including BACH2, RORγt, FOSL2, AP-1 and RUNX1, that controlled expression of the wound-healing program, and found a CD161+ Treg cell signature in Crohn's disease mucosa associated with reduced inflammation. These findings identify CD161+ Treg cells as a population involved in controlling the balance between inflammation and epithelial barrier healing in the gut.


Asunto(s)
Mucosa Intestinal/inmunología , Subfamilia B de Receptores Similares a Lectina de Células NK/inmunología , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología , Tretinoina/inmunología , Cicatrización de Heridas/inmunología , Enfermedad de Crohn/inmunología , Humanos
7.
Immunity ; 54(11): 2447-2449, 2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34758336

RESUMEN

Retinol is shuttled to myeloid cells for conversion to retinoic acid, but the receptor facilitating uptake of SAA:retinol complexes on myeloid cells is unknown. In a recent issue of Science, Bang et al. (2021) use genetic and biochemical approaches to reveal this critical receptor to be LRP1 and show that this axis is essential for intestinal innate and adaptive immune responses.


Asunto(s)
Tretinoina , Vitamina A , Proteínas Portadoras , Tretinoina/metabolismo , Vitamina A/metabolismo
8.
Nature ; 629(8013): 869-877, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38693267

RESUMEN

Airway hillocks are stratified epithelial structures of unknown function1. Hillocks persist for months and have a unique population of basal stem cells that express genes associated with barrier function and cell adhesion. Hillock basal stem cells continually replenish overlying squamous barrier cells. They exhibit dramatically higher turnover than the abundant, largely quiescent classic pseudostratified airway epithelium. Hillocks resist a remarkably broad spectrum of injuries, including toxins, infection, acid and physical injury because hillock squamous cells shield underlying hillock basal stem cells from injury. Hillock basal stem cells are capable of massive clonal expansion that is sufficient to resurface denuded airway, and eventually regenerate normal airway epithelium with each of its six component cell types. Hillock basal stem cells preferentially stratify and keratinize in the setting of retinoic acid signalling inhibition, a known cause of squamous metaplasia2,3. Here we show that mouse hillock expansion is the cause of vitamin A deficiency-induced squamous metaplasia. Finally, we identify human hillocks whose basal stem cells generate functional squamous barrier structures in culture. The existence of hillocks reframes our understanding of airway epithelial regeneration. Furthermore, we show that hillocks are one origin of 'squamous metaplasia', which is long thought to be a precursor of lung cancer.


Asunto(s)
Plasticidad de la Célula , Células Epiteliales , Regeneración , Mucosa Respiratoria , Células Madre , Animales , Femenino , Humanos , Masculino , Ratones , Células Epiteliales/citología , Células Epiteliales/patología , Metaplasia/etiología , Metaplasia/patología , Mucosa Respiratoria/citología , Mucosa Respiratoria/lesiones , Mucosa Respiratoria/patología , Células Madre/citología , Tretinoina/metabolismo , Tretinoina/farmacología , Vitamina A/metabolismo , Vitamina A/farmacología , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/patología , Ratones Endogámicos C57BL
9.
Genes Dev ; 36(7-8): 451-467, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35450883

RESUMEN

Genome organization plays a pivotal role in transcription, but how transcription factors (TFs) rewire the structure of the genome to initiate and maintain the programs that lead to oncogenic transformation remains poorly understood. Acute promyelocytic leukemia (APL) is a fatal subtype of leukemia driven by a chromosomal translocation between the promyelocytic leukemia (PML) and retinoic acid receptor α (RARα) genes. We used primary hematopoietic stem and progenitor cells (HSPCs) and leukemic blasts that express the fusion protein PML-RARα as a paradigm to temporally dissect the dynamic changes in the epigenome, transcriptome, and genome architecture induced during oncogenic transformation. We found that PML-RARα initiates a continuum of topologic alterations, including switches from A to B compartments, transcriptional repression, loss of active histone marks, and gain of repressive histone marks. Our multiomics-integrated analysis identifies Klf4 as an early down-regulated gene in PML-RARα-driven leukemogenesis. Furthermore, we characterized the dynamic alterations in the Klf4 cis-regulatory network during APL progression and demonstrated that ectopic Klf4 overexpression can suppress self-renewal and reverse the differentiation block induced by PML-RARα. Our study provides a comprehensive in vivo temporal dissection of the epigenomic and topological reprogramming induced by an oncogenic TF and illustrates how topological architecture can be used to identify new drivers of malignant transformation.


Asunto(s)
Leucemia Promielocítica Aguda , Diferenciación Celular/genética , Transformación Celular Neoplásica/genética , Humanos , Factor 4 Similar a Kruppel , Leucemia Promielocítica Aguda/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Factores de Transcripción/metabolismo , Tretinoina/farmacología
10.
Nat Immunol ; 18(6): 642-653, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28436955

RESUMEN

It remains unclear whether activated inflammatory macrophages can adopt features of tissue-resident macrophages, or what mechanisms might mediate such a phenotypic conversion. Here we show that vitamin A is required for the phenotypic conversion of interleukin 4 (IL-4)-activated monocyte-derived F4/80intCD206+PD-L2+MHCII+ macrophages into macrophages with a tissue-resident F4/80hiCD206-PD-L2-MHCII-UCP1+ phenotype in the peritoneal cavity of mice and during the formation of liver granulomas in mice infected with Schistosoma mansoni. The phenotypic conversion of F4/80intCD206+ macrophages into F4/80hiCD206- macrophages was associated with almost complete remodeling of the chromatin landscape, as well as alteration of the transcriptional profiles. Vitamin A-deficient mice infected with S. mansoni had disrupted liver granuloma architecture and increased mortality, which indicates that failure to convert macrophages from the F4/80intCD206+ phenotype to F4/80hiCD206- may lead to dysregulated inflammation during helminth infection.


Asunto(s)
Granuloma/inmunología , Hígado/inmunología , Macrófagos/inmunología , Esquistosomiasis mansoni/inmunología , Deficiencia de Vitamina A/inmunología , Animales , Antígenos de Diferenciación/metabolismo , Citometría de Flujo , Antígenos de Histocompatibilidad Clase II/metabolismo , Interleucina-4/inmunología , Lectinas Tipo C/metabolismo , Hígado/patología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Receptor de Manosa , Lectinas de Unión a Manosa/metabolismo , Ratones , Cavidad Peritoneal/citología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Superficie Celular/metabolismo , Schistosoma mansoni , Esquistosomiasis mansoni/patología , Tretinoina/farmacología , Proteína Desacopladora 1/metabolismo , Vitaminas/farmacología
11.
Cell ; 157(4): 832-44, 2014 May 08.
Artículo en Inglés | MEDLINE | ID: mdl-24792964

RESUMEN

Tissue-resident macrophages are highly heterogeneous in terms of their functions and phenotypes as a consequence of adaptation to different tissue environments. Local tissue-derived signals are thought to control functional polarization of resident macrophages; however, the identity of these signals remains largely unknown. It is also unknown whether functional heterogeneity is a result of irreversible lineage-specific differentiation or a consequence of continuous but reversible induction of diverse functional programs. Here, we identified retinoic acid as a signal that induces tissue-specific localization and functional polarization of peritoneal macrophages through the reversible induction of transcription factor GATA6. We further found that GATA6 in macrophages regulates gut IgA production through peritoneal B-1 cells. These results provide insight into the regulation of tissue-resident macrophage functional specialization by tissue-derived signals.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Regulación de la Expresión Génica , Macrófagos Peritoneales/citología , Tretinoina/metabolismo , Animales , Inmunoglobulina A/genética , Macrófagos Peritoneales/metabolismo , Ratones , Ratones Endogámicos C57BL , Epiplón/citología
12.
Nature ; 614(7948): 509-520, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36543322

RESUMEN

The segmented body plan of vertebrates is established during somitogenesis, a well-studied process in model organisms; however, the details of this process in humans remain largely unknown owing to ethical and technical limitations. Despite recent advances with pluripotent stem cell-based approaches1-5, models that robustly recapitulate human somitogenesis in both space and time remain scarce. Here we introduce a pluripotent stem cell-derived mesoderm-based 3D model of human segmentation and somitogenesis-which we termed 'axioloid'-that captures accurately the oscillatory dynamics of the segmentation clock and the morphological and molecular characteristics of sequential somite formation in vitro. Axioloids show proper rostrocaudal patterning of forming segments and robust anterior-posterior FGF-WNT signalling gradients and retinoic acid signalling components. We identify an unexpected critical role of retinoic acid signalling in the stabilization of forming segments, indicating distinct, but also synergistic effects of retinoic acid and extracellular matrix on the formation and epithelialization of somites. Comparative analysis demonstrates marked similarities of axioloids to the human embryo, further validated by the presence of a Hox code in axioloids. Finally, we demonstrate the utility of axioloids for studying the pathogenesis of human congenital spine diseases using induced pluripotent stem cells with mutations in HES7 and MESP2. Our results indicate that axioloids represent a promising platform for the study of axial development and disease in humans.


Asunto(s)
Tipificación del Cuerpo , Técnicas de Cultivo Tridimensional de Células , Somitos , Humanos , Tipificación del Cuerpo/efectos de los fármacos , Matriz Extracelular/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Técnicas In Vitro , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Modelos Biológicos , Mutación , Somitos/citología , Somitos/efectos de los fármacos , Somitos/embriología , Somitos/metabolismo , Enfermedades de la Columna Vertebral/patología , Tretinoina/metabolismo , Tretinoina/farmacología , Vía de Señalización Wnt/efectos de los fármacos
13.
Nature ; 623(7988): 792-802, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37968392

RESUMEN

Optimal tissue recovery and organismal survival are achieved by spatiotemporal tuning of tissue inflammation, contraction and scar formation1. Here we identify a multipotent fibroblast progenitor marked by CD201 expression in the fascia, the deepest connective tissue layer of the skin. Using skin injury models in mice, single-cell transcriptomics and genetic lineage tracing, ablation and gene deletion models, we demonstrate that CD201+ progenitors control the pace of wound healing by generating multiple specialized cell types, from proinflammatory fibroblasts to myofibroblasts, in a spatiotemporally tuned sequence. We identified retinoic acid and hypoxia signalling as the entry checkpoints into proinflammatory and myofibroblast states. Modulating CD201+ progenitor differentiation impaired the spatiotemporal appearances of fibroblasts and chronically delayed wound healing. The discovery of proinflammatory and myofibroblast progenitors and their differentiation pathways provide a new roadmap to understand and clinically treat impaired wound healing.


Asunto(s)
Receptor de Proteína C Endotelial , Fascia , Cicatrización de Heridas , Animales , Ratones , Diferenciación Celular , Hipoxia de la Célula , Linaje de la Célula , Modelos Animales de Enfermedad , Receptor de Proteína C Endotelial/metabolismo , Fascia/citología , Fascia/lesiones , Fascia/metabolismo , Fibroblastos/citología , Fibroblastos/metabolismo , Perfilación de la Expresión Génica , Inflamación/metabolismo , Inflamación/patología , Miofibroblastos/citología , Miofibroblastos/metabolismo , Transducción de Señal , Análisis de Expresión Génica de una Sola Célula , Piel/citología , Piel/lesiones , Piel/metabolismo , Tretinoina/metabolismo
14.
EMBO J ; 43(18): 3895-3915, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39060515

RESUMEN

Dendritic cell (DC) dysfunction is known to exacerbate intestinal pathologies, but the mechanisms compromising DC-mediated immune regulation in this context remain unclear. Here, we show that intestinal dendritic cells from a mouse model of experimental colitis exhibit significant levels of noncanonical NF-κB signaling, which activates the RelB:p52 heterodimer. Genetic inactivation of this pathway in DCs alleviates intestinal pathologies in mice suffering from colitis. Deficiency of RelB:p52 diminishes transcription of Axin1, a critical component of the ß-catenin destruction complex, reinforcing ß-catenin-dependent expression of Raldh2, which imparts tolerogenic DC attributes by promoting retinoic acid synthesis. DC-specific impairment of noncanonical NF-κB signaling leads to increased colonic numbers of Tregs and IgA+ B cells, which promote luminal IgA production and foster eubiosis. Experimentally introduced ß-catenin haploinsufficiency in DCs with deficient noncanonical NF-κB signaling moderates Raldh2 activity, reinstating colitogenic sensitivity in mice. Finally, inflammatory bowel-disease patients also display a deleterious noncanonical NF-κB signaling signature in intestinal DCs. In sum, we establish how noncanonical NF-κB signaling in dendritic cells can subvert retinoic acid synthesis to fuel intestinal inflammation.


Asunto(s)
Colitis , Células Dendríticas , FN-kappa B , Transducción de Señal , beta Catenina , Animales , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Ratones , beta Catenina/metabolismo , beta Catenina/genética , FN-kappa B/metabolismo , Colitis/inmunología , Colitis/metabolismo , Colitis/inducido químicamente , Colitis/patología , Colitis/genética , Factor de Transcripción ReIB/metabolismo , Factor de Transcripción ReIB/genética , Retinal-Deshidrogenasa/metabolismo , Retinal-Deshidrogenasa/genética , Humanos , Ratones Endogámicos C57BL , Subunidad p52 de NF-kappa B/metabolismo , Subunidad p52 de NF-kappa B/genética , Modelos Animales de Enfermedad , Ratones Noqueados , Tolerancia Inmunológica , Tretinoina/metabolismo , Aldehído Oxidorreductasas
15.
Immunity ; 50(5): 1276-1288.e5, 2019 05 21.
Artículo en Inglés | MEDLINE | ID: mdl-30902637

RESUMEN

Microbes colonize all body surfaces at birth and participate in the development of the immune system. In newborn mammals, the intestinal microbiota is first shaped by the dietary and immunological components of milk and then changes upon the introduction of solid food during weaning. Here, we explored the reactivity of the mouse intestinal immune system during the first weeks after birth and into adulthood. At weaning, the intestinal microbiota induced a vigorous immune response-a "weaning reaction"-that was programmed in time. Inhibition of the weaning reaction led to pathological imprinting and increased susceptibility to colitis, allergic inflammation, and cancer later in life. Prevention of this pathological imprinting was associated with the generation of RORγt+ regulatory T cells, which required bacterial and dietary metabolites-short-chain fatty acids and retinoic acid. Thus, the weaning reaction to microbiota is required for immune ontogeny, the perturbation of which leads to increased susceptibility to immunopathologies later in life.


Asunto(s)
Microbioma Gastrointestinal/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Linfocitos T Reguladores/inmunología , Destete , Animales , Animales Recién Nacidos/inmunología , Animales Recién Nacidos/microbiología , Ácidos Grasos Volátiles/metabolismo , Ratones , Ratones Endogámicos C57BL , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Tretinoina/metabolismo
16.
Immunity ; 51(1): 119-130.e5, 2019 07 16.
Artículo en Inglés | MEDLINE | ID: mdl-31231034

RESUMEN

Tissue-resident macrophages require specific milieus for the maintenance of defining gene-expression programs. Expression of the transcription factor GATA6 is required for the homeostasis, function and localization of peritoneal cavity-resident macrophages. Gata6 expression is maintained in a non-cell autonomous manner and is elicited by the vitamin A metabolite, retinoic acid. Here, we found that the GATA6 transcriptional program is a common feature of macrophages residing in all visceral body cavities. Retinoic acid-dependent and -independent hallmark genes of GATA6+ macrophages were induced by mesothelial and fibroblastic stromal cells that express the transcription factor Wilms' Tumor 1 (WT1), which drives the expression of two rate-limiting enzymes in retinol metabolism. Depletion of Wt1+ stromal cells reduced the frequency of GATA6+ macrophages in the peritoneal, pleural and pericardial cavities. Thus, Wt1+ mesothelial and fibroblastic stromal cells constitute essential niche components supporting the tissue-specifying transcriptional landscape and homeostasis of cavity-resident macrophages.


Asunto(s)
Factor de Transcripción GATA6/metabolismo , Macrófagos/fisiología , Pericardio/inmunología , Cavidad Peritoneal/fisiología , Cavidad Pleural/inmunología , Proteínas Represoras/metabolismo , Células del Estroma/fisiología , Animales , Diferenciación Celular , Células Cultivadas , Factor de Transcripción GATA6/genética , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Represoras/genética , Tretinoina/metabolismo , Proteínas WT1
17.
Immunity ; 50(1): 106-120.e10, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30650370

RESUMEN

CD4+ T helper (Th) differentiation is regulated by diverse inputs, including the vitamin A metabolite retinoic acid (RA). RA acts through its receptor RARα to repress transcription of inflammatory cytokines, but is also essential for Th-mediated immunity, indicating complex effects of RA on Th specification and the outcome of the immune response. We examined the impact of RA on the genome-wide transcriptional response during Th differentiation to multiple subsets. RA effects were subset-selective and were most significant in Th9 cells. RA globally antagonized Th9-promoting transcription factors and inhibited Th9 differentiation. RA directly targeted the extended Il9 locus and broadly modified the Th9 epigenome through RARα. RA-RARα activity limited murine Th9-associated pulmonary inflammation, and human allergic inflammation was associated with reduced expression of RA target genes. Thus, repression of the Th9 program is a major function of RA-RARα signaling in Th differentiation, arguing for a role for RA in interleukin 9 (IL-9) related diseases.


Asunto(s)
Hipersensibilidad/inmunología , Pulmón/fisiología , Neumonía/inmunología , Receptor alfa de Ácido Retinoico/metabolismo , Linfocitos T Colaboradores-Inductores/fisiología , Animales , Represión Epigenética , Células HEK293 , Humanos , Hipersensibilidad/genética , Interleucina-9/metabolismo , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/genética , Receptor alfa de Ácido Retinoico/genética , Transducción de Señal , Transcripción Genética , Tretinoina/metabolismo
18.
Cell ; 155(1): 215-27, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-24074870

RESUMEN

Hematopoietic stem cells (HSCs) develop from a specialized subpopulation of endothelial cells known as hemogenic endothelium (HE). Although the HE origin of HSCs is now well established in different species, the signaling pathways that control this transition remain poorly understood. Here, we show that activation of retinoic acid (RA) signaling in aorta-gonad-mesonephros-derived HE ex vivo dramatically enhanced its HSC potential, whereas conditional inactivation of the RA metabolizing enzyme retinal dehydrogenase 2 in VE-cadherin expressing endothelial cells in vivo abrogated HSC development. Wnt signaling completely blocked the HSC inductive effects of RA modulators, whereas inhibition of the pathway promoted the development of HSCs in the absence of RA signaling. Collectively, these findings position RA and Wnt signaling as key regulators of HSC development and in doing so provide molecular insights that will aid in developing strategies for their generation from pluripotent stem cells.


Asunto(s)
Células Madre Hematopoyéticas/citología , Tretinoina/metabolismo , Aldehído Oxidorreductasas/metabolismo , Animales , Aorta/citología , Aorta/embriología , Regulación hacia Abajo , Embrión de Mamíferos , Gónadas/citología , Gónadas/embriología , Células Madre Hematopoyéticas/metabolismo , Mesonefro/citología , Ratones , Receptores de Ácido Retinoico/metabolismo , Vía de Señalización Wnt
19.
Development ; 151(16)2024 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-39082789

RESUMEN

Skeletal muscles of the head and trunk originate in distinct lineages with divergent regulatory programmes converging on activation of myogenic determination factors. Branchiomeric head and neck muscles share a common origin with cardiac progenitor cells in cardiopharyngeal mesoderm (CPM). The retinoic acid (RA) signalling pathway is required during a defined early time window for normal deployment of cells from posterior CPM to the heart. Here, we show that blocking RA signalling in the early mouse embryo also results in selective loss of the trapezius neck muscle, without affecting other skeletal muscles. RA signalling is required for robust expression of myogenic determination factors in posterior CPM and subsequent expansion of the trapezius primordium. Lineage-specific activation of a dominant-negative RA receptor reveals that trapezius development is not regulated by direct RA signalling to myogenic progenitor cells in CPM, or through neural crest cells, but indirectly through the somitic lineage, closely apposed with posterior CPM in the early embryo. These findings suggest that trapezius development is dependent on precise spatiotemporal interactions between cranial and somitic mesoderm at the head/trunk interface.


Asunto(s)
Cabeza , Mesodermo , Desarrollo de Músculos , Músculos del Cuello , Transducción de Señal , Tretinoina , Animales , Tretinoina/metabolismo , Ratones , Músculos del Cuello/embriología , Mesodermo/metabolismo , Mesodermo/embriología , Cabeza/embriología , Regulación del Desarrollo de la Expresión Génica , Somitos/metabolismo , Somitos/embriología , Receptores de Ácido Retinoico/metabolismo
20.
Development ; 151(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39250350

RESUMEN

Dorsal neural tube-derived retinoic acid promotes the end of neural crest production and transition into a definitive roof plate. Here, we analyze how this impacts the segregation of central and peripheral lineages, a process essential for tissue patterning and function. Localized in ovo inhibition in quail embryos of retinoic acid activity followed by single-cell transcriptomics unraveled a comprehensive list of differentially expressed genes relevant to these processes. Importantly, progenitors co-expressed neural crest, roof plate and dI1 interneuron markers, indicating a failure in proper lineage segregation. Furthermore, separation between roof plate and dI1 interneurons is mediated by Notch activity downstream of retinoic acid, highlighting their crucial role in establishing the roof plate-dI1 boundary. Within the peripheral branch, where absence of retinoic acid resulted in neural crest production and emigration extending into the roof plate stage, sensory progenitors failed to separate from melanocytes, leading to formation of a common glia-melanocyte cell with aberrant migratory patterns. In summary, the implementation of single-cell RNA sequencing facilitated the discovery and characterization of a molecular mechanism responsible for the segregation of dorsal neural fates during development.


Asunto(s)
Cresta Neural , Tretinoina , Animales , Tretinoina/metabolismo , Tretinoina/farmacología , Cresta Neural/metabolismo , Cresta Neural/citología , Regulación del Desarrollo de la Expresión Génica , Codorniz/embriología , Movimiento Celular , Receptores Notch/metabolismo , Linaje de la Célula , Tipificación del Cuerpo/genética , Tipificación del Cuerpo/efectos de los fármacos , Placa Neural/metabolismo , Placa Neural/embriología , Interneuronas/metabolismo , Interneuronas/citología , Análisis de la Célula Individual , Tubo Neural/embriología , Tubo Neural/metabolismo , Diferenciación Celular , Melanocitos/metabolismo , Melanocitos/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA