Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Stem Cell ; 30(9): 1166-1178.e8, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37597516

RESUMO

The intestinal epithelium has high intrinsic turnover rate, and the precise renewal of the epithelium is dependent on the microenvironment. The intestine is innervated by a dense network of peripheral nerves that controls various aspects of intestinal physiology. However, the role of neurons in regulating epithelial cell regeneration remains largely unknown. Here, we investigated the effects of gut-innervating adrenergic nerves on epithelial cell repair following irradiation (IR)-induced injury. We observed that adrenergic nerve density in the small intestine increased post IR, while chemical adrenergic denervation impaired epithelial regeneration. Single-cell RNA sequencing experiments revealed a decrease in IL-22 signaling post IR in denervated animals. Combining pharmacologic and genetic tools, we demonstrate that ß-adrenergic receptor signaling drives IL-22 production from type 3 innate lymphoid cells (ILC3s) post IR, which in turn promotes epithelial regeneration. These results define an adrenergic-ILC3 axis important for intestinal regeneration.


Assuntos
Neurônios Adrenérgicos , Imunidade Inata , Mucosa Intestinal , Linfócitos , Regeneração , Animais , Transdução de Sinais , Neurônios Adrenérgicos/fisiologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/inervação , Mucosa Intestinal/fisiologia , Camundongos , Interleucina 22
2.
Blood Adv ; 7(4): 491-507, 2023 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35914228

RESUMO

Self-renewal and differentiation of stem and progenitor cells are tightly regulated to ensure tissue homeostasis. This regulation is enabled both remotely by systemic circulating cues, such as cytokines and hormones, and locally by various niche-confined factors. R-spondin 3 (RSPO3) is one of the most potent enhancers of Wnt signaling, and its expression is usually restricted to the stem cell niche where it provides localized enhancement of Wnt signaling to regulate stem cell expansion and differentiation. Disruption of this niche-confined expression can disturb proper tissue organization and lead to cancers. Here, we investigate the consequences of disrupting the niche-restricted expression of RSPO3 in various tissues, including the hematopoietic system. We show that normal Rspo3 expression is confined to the perivascular niche in the bone marrow. Induction of increased systemic levels of circulating RSPO3 outside of the niche results in prominent loss of early B-cell progenitors and anemia but surprisingly has no effect on hematopoietic stem cells. Using molecular, pharmacologic, and genetic approaches, we show that these RSPO3-induced hematopoietic phenotypes are Wnt and RSPO3 dependent and mediated through noncanonical Wnt signaling. Our study highlights a distinct role for a Wnt/RSPO3 signaling axis in the regulation of hematopoiesis, as well as possible challenges related to therapeutic use of RSPOs for regenerative medicine.


Assuntos
Hematopoese , Nicho de Células-Tronco , Hematopoese/genética , Células-Tronco Hematopoéticas , Diferenciação Celular/genética , Via de Sinalização Wnt/fisiologia
3.
Sci Immunol ; 6(59)2021 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-33963061

RESUMO

Repair of the intestinal epithelium is tightly regulated to maintain homeostasis. The response after epithelial damage needs to be local and proportional to the insult. How different types of damage are coupled to repair remains incompletely understood. We report that after distinct types of intestinal epithelial damage, IL-1R1 signaling in GREM1+ mesenchymal cells increases production of R-spondin 3 (RSPO3), a Wnt agonist required for intestinal stem cell self-renewal. In parallel, IL-1R1 signaling regulates IL-22 production by innate lymphoid cells and promotes epithelial hyperplasia and regeneration. Although the regulation of both RSPO3 and IL-22 is critical for epithelial recovery from Citrobacter rodentium infection, IL-1R1-dependent RSPO3 production by GREM1+ mesenchymal cells alone is sufficient and required for recovery after dextran sulfate sodium-induced colitis. These data demonstrate how IL-1R1-dependent signaling orchestrates distinct repair programs tailored to the type of injury sustained that are required to restore intestinal epithelial barrier function.


Assuntos
Citrobacter rodentium , Infecções por Enterobacteriaceae/imunologia , Mucosa Intestinal/fisiologia , Receptores Tipo I de Interleucina-1/imunologia , Animais , Células Cultivadas , Técnicas de Cocultura , Colite/induzido quimicamente , Colite/imunologia , Colite/patologia , Colo/efeitos dos fármacos , Colo/imunologia , Colo/patologia , Sulfato de Dextrana , Células Epiteliais , Fibroblastos , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Camundongos Transgênicos , Organoides , Receptores Tipo I de Interleucina-1/genética , Regeneração , Transdução de Sinais , Trombospondinas/imunologia , Interleucina 22
4.
Nat Immunol ; 22(5): 571-585, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33903764

RESUMO

Fibroblastic reticular cells (FRCs) are specialized stromal cells that define tissue architecture and regulate lymphocyte compartmentalization, homeostasis, and innate and adaptive immunity in secondary lymphoid organs (SLOs). In the present study, we used single-cell RNA sequencing (scRNA-seq) of human and mouse lymph nodes (LNs) to identify a subset of T cell-zone FRCs defined by the expression of Gremlin1 (Grem1) in both species. Grem1-CreERT2 knock-in mice enabled localization, multi-omics characterization and genetic depletion of Grem1+ FRCs. Grem1+ FRCs primarily localize at T-B cell junctions of SLOs, neighboring pre-dendritic cells and conventional dendritic cells (cDCs). As such, their depletion resulted in preferential loss and decreased homeostatic proliferation and survival of resident cDCs and compromised T cell immunity. Trajectory analysis of human LN scRNA-seq data revealed expression similarities to murine FRCs, with GREM1+ cells marking the endpoint of both trajectories. These findings illuminate a new Grem1+ fibroblastic niche in LNs that functions to maintain the homeostasis of lymphoid tissue-resident cDCs.


Assuntos
Células Dendríticas Foliculares/imunologia , Fibroblastos/imunologia , Linfonodos/imunologia , Células Estromais/imunologia , Idoso , Animais , Apoptose/genética , Apoptose/imunologia , Proliferação de Células/genética , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Células Dendríticas Foliculares/metabolismo , Feminino , Fibroblastos/metabolismo , Regulação da Expressão Gênica/imunologia , Técnicas de Introdução de Genes , Humanos , Imunidade Celular/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Linfonodos/citologia , Masculino , Camundongos , Camundongos Transgênicos , RNA-Seq , Análise de Célula Única , Células Estromais/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
5.
Cell Stem Cell ; 26(3): 391-402.e5, 2020 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-32084389

RESUMO

Intestinal stem cells (ISCs) are confined to crypt bottoms and their progeny differentiate near crypt-villus junctions. Wnt and bone morphogenic protein (BMP) gradients drive this polarity, and colorectal cancer fundamentally reflects disruption of this homeostatic signaling. However, sub-epithelial sources of crucial agonists and antagonists that organize this BMP gradient remain obscure. Here, we couple whole-mount high-resolution microscopy with ensemble and single-cell RNA sequencing (RNA-seq) to identify three distinct PDGFRA+ mesenchymal cell types. PDGFRA(hi) telocytes are especially abundant at the villus base and provide a BMP reservoir, and we identified a CD81+ PDGFRA(lo) population present just below crypts that secretes the BMP antagonist Gremlin1. These cells, referred to as trophocytes, are sufficient to expand ISCs in vitro without additional trophic support and contribute to ISC maintenance in vivo. This study reveals intestinal mesenchymal structure at fine anatomic, molecular, and functional detail and the cellular basis for a signaling gradient necessary for tissue self-renewal.


Assuntos
Intestinos , Transdução de Sinais , Proliferação de Células , Mucosa Intestinal , Células-Tronco
6.
Nature ; 543(7647): 676-680, 2017 03 29.
Artigo em Inglês | MEDLINE | ID: mdl-28358093

RESUMO

Cancer stem cells (CSCs) have been hypothesized to represent the driving force behind tumour progression and metastasis, making them attractive cancer targets. However, conclusive experimental evidence for their functional relevance is still lacking for most malignancies. Here we show that the leucine-rich repeat-containing G-protein-coupled receptor 5 (Lgr5) identifies intestinal CSCs in mouse tumours engineered to recapitulate the clinical progression of human colorectal cancer. We demonstrate that selective Lgr5+ cell ablation restricts primary tumour growth, but does not result in tumour regression. Instead, tumours are maintained by proliferative Lgr5- cells that continuously attempt to replenish the Lgr5+ CSC pool, leading to rapid re-initiation of tumour growth upon treatment cessation. Notably, CSCs are critical for the formation and maintenance of liver metastasis derived from colorectal cancers. Together, our data highlight distinct CSC dependencies for primary versus metastasic tumour growth, and suggest that targeting CSCs may represent a therapeutic opportunity for managing metastatic disease.


Assuntos
Neoplasias Colorretais/patologia , Metástase Neoplásica/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Receptores Acoplados a Proteínas G/metabolismo , Animais , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/metabolismo , Proliferação de Células , Separação Celular , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Modelos Animais de Doenças , Progressão da Doença , Feminino , Injeções Subcutâneas , Intestinos/patologia , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/patologia , Neoplasias Hepáticas/secundário , Camundongos , Metástase Neoplásica/tratamento farmacológico , Células-Tronco Neoplásicas/efeitos dos fármacos , Organoides/patologia , Organoides/transplante , Receptores Acoplados a Proteínas G/análise
7.
Nature ; 529(7584): 97-100, 2016 Jan 07.
Artigo em Inglês | MEDLINE | ID: mdl-26700806

RESUMO

Colorectal cancer remains a major unmet medical need, prompting large-scale genomics efforts in the field to identify molecular drivers for which targeted therapies might be developed. We previously reported the identification of recurrent translocations in R-spondin genes present in a subset of colorectal tumours. Here we show that targeting RSPO3 in PTPRK-RSPO3-fusion-positive human tumour xenografts inhibits tumour growth and promotes differentiation. Notably, genes expressed in the stem-cell compartment of the intestine were among those most sensitive to anti-RSPO3 treatment. This observation, combined with functional assays, suggests that a stem-cell compartment drives PTPRK-RSPO3 colorectal tumour growth and indicates that the therapeutic targeting of stem-cell properties within tumours may be a clinically relevant approach for the treatment of colorectal tumours.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/patologia , Terapia de Alvo Molecular , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Proteínas Tirosina Fosfatases Classe 2 Semelhantes a Receptores/metabolismo , Trombospondinas/metabolismo , Animais , Anticorpos/imunologia , Anticorpos/farmacologia , Anticorpos/uso terapêutico , Divisão Celular/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Progressão da Doença , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Mucosa Intestinal/metabolismo , Intestinos/citologia , Intestinos/efeitos dos fármacos , Intestinos/patologia , Masculino , Camundongos , Células-Tronco Neoplásicas/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Trombospondinas/antagonistas & inibidores , Trombospondinas/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Nature ; 488(7413): 660-4, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22895193

RESUMO

Identifying and understanding changes in cancer genomes is essential for the development of targeted therapeutics. Here we analyse systematically more than 70 pairs of primary human colon tumours by applying next-generation sequencing to characterize their exomes, transcriptomes and copy-number alterations. We have identified 36,303 protein-altering somatic changes that include several new recurrent mutations in the Wnt pathway gene TCF7L2, chromatin-remodelling genes such as TET2 and TET3 and receptor tyrosine kinases including ERBB3. Our analysis for significantly mutated cancer genes identified 23 candidates, including the cell cycle checkpoint kinase ATM. Copy-number and RNA-seq data analysis identified amplifications and corresponding overexpression of IGF2 in a subset of colon tumours. Furthermore, using RNA-seq data we identified multiple fusion transcripts including recurrent gene fusions involving R-spondin family members RSPO2 and RSPO3 that together occur in 10% of colon tumours. The RSPO fusions were mutually exclusive with APC mutations, indicating that they probably have a role in the activation of Wnt signalling and tumorigenesis. Consistent with this we show that the RSPO fusion proteins were capable of potentiating Wnt signalling. The R-spondin gene fusions and several other gene mutations identified in this study provide new potential opportunities for therapeutic intervention in colon cancer.


Assuntos
Neoplasias do Colo/genética , Fusão Gênica/genética , Genes Neoplásicos/genética , Peptídeos e Proteínas de Sinalização Intercelular/genética , Trombospondinas/genética , Proteínas Mutadas de Ataxia Telangiectasia , Sequência de Bases , Proteínas de Ciclo Celular/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Variações do Número de Cópias de DNA/genética , Proteínas de Ligação a DNA/genética , Dioxigenases/genética , Exoma/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Genes APC , Humanos , Fator de Crescimento Insulin-Like II/genética , Dados de Sequência Molecular , Mutação/genética , Polimorfismo de Nucleotídeo Único/genética , Proteínas Serina-Treonina Quinases/genética , Proteínas Proto-Oncogênicas/genética , Receptor ErbB-3/genética , Análise de Sequência de RNA , Transdução de Sinais/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Proteínas Wnt/metabolismo
9.
Development ; 133(9): 1831-44, 2006 May.
Artigo em Inglês | MEDLINE | ID: mdl-16613831

RESUMO

Mouse embryos bearing hypomorphic and conditional null Fgf8 mutations have small and abnormally patterned telencephalons. We provide evidence that the hypoplasia results from decreased Foxg1 expression, reduced cell proliferation and increased cell death. In addition, alterations in the expression of Bmp4, Wnt8b, Nkx2.1 and Shh are associated with abnormal development of dorsal and ventral structures. Furthermore, nonlinear effects of Fgf8 gene dose on the expression of a subset of genes, including Bmp4 and Msx1, correlate with a holoprosencephaly phenotype and with the nonlinear expression of transcription factors that regulate neocortical patterning. These data suggest that Fgf8 functions to coordinate multiple patterning centers, and that modifications in the relative strength of FGF signaling can have profound effects on the relative size and nature of telencephalic subdivisions.


Assuntos
Padronização Corporal , Fator 8 de Crescimento de Fibroblasto/genética , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Telencéfalo/embriologia , Alelos , Animais , Apoptose , Proliferação de Células , Fator 8 de Crescimento de Fibroblasto/metabolismo , Camundongos , Camundongos Mutantes , Modelos Biológicos , Mutação , Telencéfalo/citologia
10.
Neuron ; 47(4): 483-6, 2005 Aug 18.
Artigo em Inglês | MEDLINE | ID: mdl-16102531

RESUMO

Mammals have developed patterns of social relationships that enhance the survival of individuals and maximize the reproductive success of species. Although social stimuli and social responses are highly complex, recent studies are providing substantial insights into their neural substrates. Neural pathways employing the nonapeptides vasopressin and oxytocin play a particularly prominent role both in social recognition and the expression of appropriate social responses. New insights into social neuroscience are discussed, along with the relevance of this rapidly developing field to human relationships and disease processes.


Assuntos
Encéfalo/fisiologia , Sistema Hipotálamo-Hipofisário/fisiologia , Vias Neurais/fisiologia , Neuropeptídeos/metabolismo , Comportamento Social , Animais , Humanos , Ocitocina/metabolismo , Vasopressinas/metabolismo
11.
Proc Natl Acad Sci U S A ; 100(4): 1757-62, 2003 Feb 18.
Artigo em Inglês | MEDLINE | ID: mdl-12574514

RESUMO

FGF8 is known to be an important regulator of forebrain development. Here, we investigated the effects of varying the level of Fgf8 expression in the mouse forebrain. We detected two distinct responses, one that was proportionate with Fgf8 expression and another that was not. The latter response, which led to effects on cell survival, displayed a paradoxical relationship to Fgf8 dosage. Either eliminating or increasing Fgf8 expression increased apoptosis, whereas reducing Fgf8 expression had the opposite effect. To explain these counterintuitive observations, we suggest that an FGF8-dependent cell-survival pathway is negatively regulated by intracellular inhibitors produced in proportion to FGF8 concentration. Our data provide insight into the function of FGF8 in forebrain development and underscore the value of using multiple alleles and different experimental approaches to unravel the complexities of gene function in vertebrate development.


Assuntos
Sobrevivência Celular/genética , Fatores de Crescimento de Fibroblastos/genética , Dosagem de Genes , Prosencéfalo/metabolismo , Animais , Fator 8 de Crescimento de Fibroblasto , Expressão Gênica , Genótipo , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA