Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3018, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589357

RESUMO

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced acute GI syndrome. Through single-cell RNA-sequencing of the irradiated mouse small intestine, we find that p53 target genes are specifically enriched in regenerating epithelial cells that undergo fetal-like reversion, including revival stem cells (revSCs) that promote animal survival after severe damage of the GI tract. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce fetal-like revSCs. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells and is controlled by an Mdm2-mediated negative feedback loop. Together, our findings reveal that p53 suppresses severe radiation-induced GI injury by promoting fetal-like reprogramming of irradiated intestinal epithelial cells.


Assuntos
Lesões por Radiação , Proteína Supressora de Tumor p53 , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Intestinos , Trato Gastrointestinal/metabolismo , Lesões por Radiação/genética , Lesões por Radiação/metabolismo , Células-Tronco/metabolismo , Apoptose/genética
3.
bioRxiv ; 2023 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-37162959

RESUMO

Ionizing radiation induces cell death in the gastrointestinal (GI) epithelium by activating p53. However, p53 also prevents animal lethality caused by radiation-induced GI injury. Through single-cell RNA-sequencing of the irradiated mouse intestine, we find that p53 target genes are specifically enriched in stem cells of the regenerating epithelium, including revival stem cells that promote animal survival after GI damage. Accordingly, in mice with p53 deleted specifically in the GI epithelium, ionizing radiation fails to induce revival stem cells. Using intestinal organoids, we show that transient p53 expression is required for the induction of revival stem cells that is controlled by an Mdm2-mediated negative feedback loop. These results suggest that p53 suppresses severe radiation-indued GI injury by promoting intestinal epithelial cell reprogramming. One-Sentence Summary: After severe radiation injury to the intestine, transient p53 activity induces revival stem cells to promote regeneration.

4.
Nat Commun ; 14(1): 3150, 2023 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-37258521

RESUMO

How the genetic landscape governs a tumor's response to immunotherapy remains poorly understood. To assess the immune-modulatory capabilities of 573 genes associated with altered cytotoxicity in human cancers, here we perform CRISPR/Cas9 screens directly in mouse lung cancer models. We recover the known immune evasion factors Stat1 and Serpinb9 and identify the cancer testis antigen Adam2 as an immune modulator, whose expression is induced by KrasG12D and further elevated by immunotherapy. Using loss- and gain-of-function experiments, we show that ADAM2 functions as an oncogene by restraining interferon and TNF cytokine signaling causing reduced presentation of tumor-associated antigens. ADAM2 also restricts expression of the immune checkpoint inhibitors PDL1, LAG3, TIGIT and TIM3 in the tumor microenvironment, which might explain why ex vivo expanded and adoptively transferred cytotoxic T-cells show enhanced cytotoxic efficacy in ADAM2 overexpressing tumors. Together, direct in vivo CRISPR/Cas9 screens can uncover genetic alterations that control responses to immunotherapies.


Assuntos
Antineoplásicos , Fertilinas , Neoplasias Pulmonares , Serpinas , Animais , Humanos , Masculino , Camundongos , Antígenos de Neoplasias , Fertilinas/genética , Imunoterapia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Proteínas de Membrana/genética , Serpinas/genética , Linfócitos T Citotóxicos , Microambiente Tumoral
5.
Front Microbiol ; 13: 922727, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36081793

RESUMO

Aim: The aim was to evaluate the impact of intermittent fasting (IF) on human body mass index (BMI) and serum lipid profile thorough constructive rectification of gut microbiota. Methods and results: Fourteen healthy women and thirty-one men were included in the study. Their blood and fecal samples were collected before and at the end of the study. Blood parameters, anthropometric values, and gut microbiology were noted to investigate the impact of intermittent fasting (IF) on human gut microbiota and physiology. Our data revealed that IF reduces the body weight and improves blood lipid profile, such as increasing high-density lipoprotein (HDL) and decreasing total cholesterol, triglycerides, and low- and very low-density lipoprotein levels. IF also decreases culturable aerobic bacterial count and increased fungal count. It was also found that the gut metagenome is altered considerably after IF. The human fecal bacterial diversity exhibited significant changes in decreased overall bacterial population, increased bacterial diversity (alpha diversity), and promoted evenness within the bacterial population at the species level. Anti-inflammatory bacteria Lactobacillus and Bifidobacterium were favorably increased, while pathogenic bacteria were decreased. Conclusion: Collectively, these results indicated that IF could improve lipid profile and body weight in humans, and the potential mechanisms might be via regulating gut microbiota. Significance and impact of the study: We demonstrated for the first time that IF improved body weight and blood lipid profile, indicating that IF could mitigate gut microbiota in humans.

6.
Gut Microbes ; 14(1): 2108281, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35939622

RESUMO

The small intestinal epithelial barrier inputs signals from the gut microbiota in order to balance physiological inflammation and tolerance, and to promote homeostasis. Understanding the dynamic relationship between microbes and intestinal epithelial cells has been a challenge given the cellular heterogeneity associated with the epithelium and the inherent difficulty of isolating and identifying individual cell types. Here, we used single-cell RNA sequencing of small intestinal epithelial cells from germ-free and specific pathogen-free mice to study microbe-epithelium crosstalk at the single-cell resolution. The presence of microbiota did not impact overall cellular composition of the epithelium, except for an increase in Paneth cell numbers. Contrary to expectations, pattern recognition receptors and their adaptors were not induced by the microbiota but showed concentrated expression in a small proportion of epithelial cell subsets. The presence of the microbiota induced the expression of host defense- and glycosylation-associated genes in distinct epithelial cell compartments. Moreover, the microbiota altered the metabolic gene expression profile of epithelial cells, consequently inducing mTOR signaling thereby suggesting microbe-derived metabolites directly activate and regulate mTOR signaling. Altogether, these findings present a resource of the homeostatic transcriptional and cellular impact of the microbiota on the small intestinal epithelium.


Assuntos
Microbioma Gastrointestinal , Animais , Mucosa Intestinal/metabolismo , Intestino Delgado , Camundongos , Celulas de Paneth , Serina-Treonina Quinases TOR/metabolismo
7.
Sci Rep ; 12(1): 3068, 2022 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-35197500

RESUMO

Serratia marcescens is an opportunistic bacterium that infects a wide range of hosts including humans. It is a potent pathogen in a septic injury model of Drosophila melanogaster since a few bacteria directly injected in the body cavity kill the insect within a day. In contrast, flies do not succumb to ingested bacteria for days even though some bacteria cross the intestinal barrier into the hemolymph within hours. The mechanisms by which S. marcescens attacks enterocytes and damages the intestinal epithelium remain uncharacterized. To better understand intestinal infections, we performed a genetic screen for loss of virulence of ingested S. marcescens and identified FliR, a structural component of the flagellum, as a virulence factor. Next, we compared the virulence of two flagellum mutants fliR and flhD in two distinct S. marcescens strains. Both genes are required for S. marcescens to escape the gut lumen into the hemocoel, indicating that the flagellum plays an important role for the passage of bacteria through the intestinal barrier. Unexpectedly, fliR but not flhD is involved in S. marcescens-mediated damages of the intestinal epithelium that ultimately contribute to the demise of the host. Our results therefore suggest a flagellum-independent role for fliR in bacterial virulence.


Assuntos
Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Drosophila melanogaster/microbiologia , Flagelos/genética , Flagelos/fisiologia , Gastroenterite/microbiologia , Mucosa Intestinal/microbiologia , Proteínas de Membrana/genética , Proteínas de Membrana/fisiologia , Infecções por Serratia , Serratia marcescens/genética , Serratia marcescens/patogenicidade , Animais , Modelos Animais de Doenças , Mucosa Intestinal/patologia , Mutação , Virulência/genética
8.
Cell Stem Cell ; 29(1): 86-100.e6, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34727519

RESUMO

The high turnover and regenerative capacity of the adult intestine relies on resident stem cells located at the bottom of the crypt. The enteric nervous system consists of an abundant network of enteric glial cells (EGCs) and neurons. Despite the close proximity of EGCs to stem cells, their in vivo role as a stem cell niche is still unclear. By analyzing the mouse and human intestinal mucosa transcriptomes at the single-cell level, we defined the regulation of EGC heterogeneity in homeostasis and chronic inflammatory bowel disease. Ablation of EGC subpopulations revealed that the repair potential of intestinal stem cells (ISCs) is regulated by a specific subset of glial fibrillary acidic protein (GFAP)+ EGCs. Mechanistically, injury induces expansion of GFAP+ EGCs, which express several WNT ligands to promote LGR5+ ISC self-renewal. Our work reveals the dynamically regulated heterogeneity of EGCs as a key part of the intestinal stem cell niche in regeneration and disease.


Assuntos
Sistema Nervoso Entérico , Nicho de Células-Tronco , Animais , Mucosa Intestinal , Intestinos , Camundongos , Neuroglia
9.
Nat Commun ; 11(1): 2591, 2020 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-32444641

RESUMO

The intestine is a highly dynamic environment that requires tight control of the various inputs to maintain homeostasis and allow for proper responses to injury. It was recently found that the stem cell niche and epithelium is regenerated after injury by de-differentiated adult cells, through a process that gives rise to Sca1+ fetal-like cells and is driven by a transient population of Clu+ revival stem cells (revSCs). However, the molecular mechanisms that regulate this dynamic process have not been fully defined. Here we show that TNFAIP8 (also known as TIPE0) is a regulator of intestinal homeostasis that is vital for proper regeneration. TIPE0 functions through inhibiting basal Akt activation by the commensal microbiota via modulating membrane phospholipid abundance. Loss of TIPE0 in mice results in injury-resistant enterocytes, that are hyperproliferative, yet have regenerative deficits and are shifted towards a de-differentiated state. Tipe0-/- enterocytes show basal induction of the Clu+ regenerative program and a fetal gene expression signature marked by Sca1, but upon injury are unable to generate Sca-1+/Clu+ revSCs and could not regenerate the epithelium. This work demonstrates the role of TIPE0 in regulating the dynamic signaling that determines the injury response and enables intestinal epithelial cell regenerative plasticity.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Microbioma Gastrointestinal/fisiologia , Intestinos/citologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Proteínas Reguladoras de Apoptose/genética , Ataxina-1/metabolismo , Diferenciação Celular , Colite/induzido quimicamente , Colite/patologia , Enterócitos/patologia , Feminino , Técnicas de Silenciamento de Genes , Homeostase , Intestinos/irrigação sanguínea , Intestinos/patologia , Intestinos/efeitos da radiação , Isquemia/genética , Isquemia/patologia , Antígenos Comuns de Leucócito/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Lesões Experimentais por Radiação/patologia , Regeneração/fisiologia , Transdução de Sinais , Nicho de Células-Tronco , Células-Tronco/metabolismo
10.
Nature ; 569(7754): 121-125, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-31019301

RESUMO

The turnover of the intestinal epithelium is driven by multipotent LGR5+ crypt-base columnar cells (CBCs) located at the bottom of crypt zones1. However, CBCs are lost following injury, such as irradiation2, but the intestinal epithelium is nevertheless able to recover3. Thus, a second population of quiescent '+4' cells, or reserve stem cells (RSCs), has previously been proposed to regenerate the damaged intestine4-7. Although CBCs and RSCs were thought to be mutually exclusive4,8, subsequent studies have found that LGR5+ CBCs express RSC markers9 and that RSCs were dispensable-whereas LGR5+ cells were essential-for repair of the damaged intestine3. In addition, progenitors of absorptive enterocytes10, secretory cells11-15 and slow cycling LGR5+ cells16 have been shown to contribute to regeneration whereas the transcriptional regulator YAP1, which is important for intestinal regeneration, was suggested to induce a pro-survival phenotype in LGR5+ cells17. Thus, whether cellular plasticity or distinct cell populations are critical for intestinal regeneration remains unknown. Here we applied single-cell RNA sequencing to profile the regenerating mouse intestine and identified a distinct, damage-induced quiescent cell type that we term the revival stem cell (revSC). revSCs are marked by high clusterin expression and are extremely rare under homoeostatic conditions, yet give rise-in a temporal hierarchy-to all the major cell types of the intestine, including LGR5+ CBCs. After intestinal damage by irradiation, targeted ablation of LGR5+ CBCs, or treatment with dextran sodium sulfate, revSCs undergo a YAP1-dependent transient expansion, reconstitute the LGR5+ CBC compartment and are required to regenerate a functional intestine. These studies thus define a unique stem cell that is mobilized by damage to revive the homoeostatic stem cell compartment and regenerate the intestinal epithelium.


Assuntos
Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Regeneração/genética , Análise de Célula Única , Células-Tronco/citologia , Células-Tronco/metabolismo , Transcriptoma , Animais , Feminino , Homeostase , Masculino , Camundongos , Camundongos Transgênicos , Células-Tronco Multipotentes/citologia , Células-Tronco Multipotentes/metabolismo , Regeneração/fisiologia , Análise de Sequência de RNA
11.
Cell Rep ; 20(11): 2527-2537, 2017 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-28903034

RESUMO

Sophisticated mechanisms that preserve genome integrity are critical to ensure the maintenance of regenerative capacity while preventing transformation of somatic stem cells (SCs), yet little is known about mechanisms regulating genome maintenance in these cells. Here, we show that intestinal stem cells (ISCs) induce the Argonaute family protein Piwi in response to JAK/STAT signaling during acute proliferative episodes. Piwi function is critical to ensure heterochromatin maintenance, suppress retrotransposon activation, and prevent DNA damage in homeostasis and under regenerative pressure. Accordingly, loss of Piwi results in the loss of actively dividing ISCs and their progenies by apoptosis. We further show that Piwi expression is sufficient to allay age-related retrotransposon expression, DNA damage, apoptosis, and mis-differentiation phenotypes in the ISC lineage, improving epithelial homeostasis. Our data identify a role for Piwi in the regulation of somatic SC function, and they highlight the importance of retrotransposon control in somatic SC maintenance.


Assuntos
Células-Tronco Adultas/citologia , Células-Tronco Adultas/metabolismo , Proteínas Argonautas/metabolismo , Senescência Celular , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/metabolismo , Animais , Apoptose , Núcleo Celular/metabolismo , Reparo do DNA , Elementos de DNA Transponíveis/genética , Perfilação da Expressão Gênica , Inativação Gênica , Heterocromatina/metabolismo , Intestinos/citologia , Janus Quinases/metabolismo , Fatores de Transcrição STAT/metabolismo
12.
F1000Res ; 6: 749, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28649369

RESUMO

The appearance of the first animal species on earth coincides with the emergence of transforming growth factor ß (TGFß) pathways. The evolution of these animals into more complex organisms coincides with a progressively increased TGFß repertoire through gene duplications and divergence, making secreted TGFß molecules the largest family of morphogenetic proteins in humans. It is therefore not surprising that TGFß pathways govern numerous aspects of human biology from early embryonic development to regeneration, hematopoiesis, neurogenesis, and immunity. Such heavy reliance on these pathways is reflected in the susceptibility to minor perturbations in pathway components that can lead to dysregulated signaling and a diverse range of human pathologies such as cancer, fibrosis, and developmental disorders. Attempts to comprehensively resolve these signaling cascades are complicated by the long-recognized paradoxical role the pathway plays in cell biology. Recently, several groups have probed examples of the disparate aspects of TGFß biology in a variety of animal models and uncovered novel context-dependent regulatory mechanisms. Here, we briefly review recent advancements and discuss their overall impact in directing future TGFß research.

13.
Nat Cell Biol ; 17(6): 736-48, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-26005834

RESUMO

Coordination of stem cell activity with inflammatory responses is critical for regeneration and homeostasis of barrier epithelia. The temporal sequence of cell interactions during injury-induced regeneration is only beginning to be understood. Here we show that intestinal stem cells (ISCs) are regulated by macrophage-like haemocytes during the early phase of regenerative responses of the Drosophila intestinal epithelium. On tissue damage, haemocytes are recruited to the intestine and secrete the BMP homologue DPP, inducing ISC proliferation by activating the type I receptor Saxophone and the Smad homologue SMOX. Activated ISCs then switch their response to DPP by inducing expression of Thickveins, a second type I receptor that has previously been shown to re-establish ISC quiescence by activating MAD. The interaction between haemocytes and ISCs promotes infection resistance, but also contributes to the development of intestinal dysplasia in ageing flies. We propose that similar interactions influence pathologies such as inflammatory bowel disease and colorectal cancer in humans.


Assuntos
Drosophila melanogaster/citologia , Hemócitos/citologia , Mucosa Intestinal/citologia , Células-Tronco/citologia , Junções Íntimas/imunologia , Transporte Ativo do Núcleo Celular , Envelhecimento/imunologia , Animais , Animais Geneticamente Modificados , Proliferação de Células , Células Cultivadas , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteínas de Drosophila/biossíntese , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Ativação Enzimática/imunologia , Receptores ErbB/metabolismo , Hemócitos/metabolismo , Hemócitos/transplante , Tolerância Imunológica/imunologia , Inflamação/imunologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Janus Quinases/metabolismo , Expectativa de Vida , Macrófagos/imunologia , Proteínas Serina-Treonina Quinases/biossíntese , Receptores de Superfície Celular/biossíntese , Receptores de Peptídeos de Invertebrados/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Regeneração/imunologia , Proteínas Smad Reguladas por Receptor/metabolismo , Células-Tronco/imunologia , Células-Tronco/metabolismo
14.
Artigo em Inglês | MEDLINE | ID: mdl-24380076

RESUMO

As a barrier epithelium, the intestinal epithelium has to coordinate physiological functions like digestion and nutrient resorption with the control of commensal bacteria and the prevention of pathogenic infections. It can therefore mount powerful innate immune and inflammatory responses, while, at the same time, maintaining tissue homeostasis through regenerative processes. How these different functions are coordinated remains unclear, and further insight is required to understand the age-related loss of homeostasis in this system, as well as the etiology of inflammatory and proliferative diseases of the gut. Recent work in Drosophila melanogaster has provided important new insight into the regulation of regenerative activity, innate immune homeostasis, commensal control, as well as age-related dysfunction in the intestine. Interestingly, many of the identified processes and mechanisms mirror similar homeostatic processes in the vertebrate intestine. This review summarized the current understanding of how innate immune responses, changes in commensal bacteria, and other challenges influence regenerative activity in the aging intestinal epithelium of flies and draws parallels to similar processes in mammals.


Assuntos
Drosophila melanogaster/microbiologia , Drosophila melanogaster/fisiologia , Envelhecimento/imunologia , Animais , Drosophila melanogaster/imunologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/patologia , Imunidade Inata , Inflamação/microbiologia , Inflamação/patologia , Modelos Animais
15.
Immunobiology ; 218(4): 635-44, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23083631

RESUMO

Drosophila melanogaster is a useful model to investigate mucosal immunity. The immune response to intestinal infections is mediated partly by the Immune deficiency (IMD) pathway, which only gets activated by a type of peptidoglycan lacking in several medically important Gram-positive bacterial species such as Staphylococcus. Thus, the intestinal host defense against such bacterial strains remains poorly known. Here, we have used Staphylococcus xylosus to develop a model of intestinal infections by Gram-positive bacteria. S. xylosus behaves as an opportunistic pathogen in a septic injury model, being able to kill only flies immunodeficient either for the Toll pathway or the cellular response. When ingested, it is controlled by IMD-independent host intestinal defenses, yet flies eventually die. Having excluded an overreaction of the immune response and the action of toxins, we find that flies actually succumb to starvation, likely as a result of a competition for sucrose between the bacteria and the flies. Fat stores of wild-type flies are severely reduced within a day, a period when sucrose is not yet exhausted in the feeding solution. Interestingly, the Toll pathway mutant MyD88 is more resistant to the ingestion of S. xylosus and to starvation than wild-type flies. MyD88 flies do not rapidly deplete their fat stores when starved, in contrast to wild-type flies. Thus, we have uncovered a novel function of MyD88 in the regulation of metabolism that appears to be independent of its known roles in immunity and development.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/imunologia , Antígenos de Diferenciação/imunologia , Proteínas de Drosophila/imunologia , Imunidade Inata , Imunidade nas Mucosas , Enteropatias/imunologia , Receptores Imunológicos/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus/imunologia , Inanição/imunologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Antígenos de Diferenciação/genética , Antígenos de Diferenciação/metabolismo , Modelos Animais de Doenças , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster , Enteropatias/genética , Enteropatias/metabolismo , Enteropatias/microbiologia , Enteropatias/patologia , Mutação , Receptores Imunológicos/genética , Receptores Imunológicos/metabolismo , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/metabolismo , Infecções Estafilocócicas/patologia , Inanição/genética , Inanição/metabolismo , Inanição/microbiologia , Inanição/patologia , Receptores Toll-Like/genética , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...