Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 147
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Genes Dev ; 35(3-4): 199-211, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33526586

RESUMO

Stem cells maintain tissues by balancing self-renewal with differentiation. A stem cell's local microenvironment, or niche, informs stem cell behavior and receives inputs at multiple levels. Increasingly, it is becoming clear that the overall metabolic status of an organism or metabolites themselves can function as integral members of the niche to alter stem cell fate. Macroscopic dietary interventions such as caloric restriction, the ketogenic diet, and a high-fat diet systemically alter an organism's metabolic state in different ways. Intriguingly, however, they all converge on a propensity to enhance self-renewal. Here, we highlight our current knowledge on how dietary changes feed into stem cell behavior across a wide variety of tissues and illuminate possible explanations for why diverse interventions can result in similar stem cell phenotypes. In so doing, we hope to inspire new avenues of inquiry into the importance of metabolism in stem cell homeostasis and disease.


Assuntos
Dieta , Células-Tronco/fisiologia , Animais , Dieta Hiperlipídica/efeitos adversos , Jejum/fisiologia , Glucose/metabolismo , Humanos , Metabolismo dos Lipídeos/fisiologia , Células-Tronco/microbiologia , Células-Tronco/parasitologia , Estresse Fisiológico/fisiologia
2.
Development ; 148(5)2021 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-33593820

RESUMO

Microbial factors influence homeostatic and oncogenic growth in the intestinal epithelium. However, we know little about immediate effects of commensal bacteria on stem cell division programs. In this study, we examined the effects of commensal Lactobacillus species on homeostatic and tumorigenic stem cell proliferation in the female Drosophila intestine. We identified Lactobacillus brevis as a potent stimulator of stem cell divisions. In a wild-type midgut, L.brevis activates growth regulatory pathways that drive stem cell divisions. In a Notch-deficient background, L.brevis-mediated proliferation causes rapid expansion of mutant progenitors, leading to accumulation of large, multi-layered tumors throughout the midgut. Mechanistically, we showed that L.brevis disrupts expression and subcellular distribution of progenitor cell integrins, supporting symmetric divisions that expand intestinal stem cell populations. Collectively, our data emphasize the impact of commensal microbes on division and maintenance of the intestinal progenitor compartment.


Assuntos
Adesão Celular , Proliferação de Células , Drosophila/metabolismo , Intestinos/citologia , Levilactobacillus brevis/fisiologia , Células-Tronco/metabolismo , Animais , Animais Geneticamente Modificados/metabolismo , Antibacterianos/farmacologia , Diferenciação Celular , Linhagem da Célula , Transformação Celular Neoplásica/efeitos dos fármacos , Regulação para Baixo , Drosophila/microbiologia , Proteínas de Drosophila/deficiência , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Integrinas/metabolismo , Intestinos/microbiologia , Levilactobacillus brevis/efeitos dos fármacos , Receptores Notch/deficiência , Receptores Notch/genética , Células-Tronco/citologia , Células-Tronco/microbiologia
3.
Proc Natl Acad Sci U S A ; 117(14): 8064-8073, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32198200

RESUMO

Gastrointestinal infections often induce epithelial damage that must be repaired for optimal gut function. While intestinal stem cells are critical for this regeneration process [R. C. van der Wath, B. S. Gardiner, A. W. Burgess, D. W. Smith, PLoS One 8, e73204 (2013); S. Kozar et al., Cell Stem Cell 13, 626-633 (2013)], how they are impacted by enteric infections remains poorly defined. Here, we investigate infection-mediated damage to the colonic stem cell compartment and how this affects epithelial repair and recovery from infection. Using the pathogen Clostridioides difficile, we show that infection disrupts murine intestinal cellular organization and integrity deep into the epithelium, to expose the otherwise protected stem cell compartment, in a TcdB-mediated process. Exposure and susceptibility of colonic stem cells to intoxication compromises their function during infection, which diminishes their ability to repair the injured epithelium, shown by altered stem cell signaling and a reduction in the growth of colonic organoids from stem cells isolated from infected mice. We also show, using both mouse and human colonic organoids, that TcdB from epidemic ribotype 027 strains does not require Frizzled 1/2/7 binding to elicit this dysfunctional stem cell state. This stem cell dysfunction induces a significant delay in recovery and repair of the intestinal epithelium of up to 2 wk post the infection peak. Our results uncover a mechanism by which an enteric pathogen subverts repair processes by targeting stem cells during infection and preventing epithelial regeneration, which prolongs epithelial barrier impairment and creates an environment in which disease recurrence is likely.


Assuntos
Proteínas de Bactérias/metabolismo , Toxinas Bacterianas/metabolismo , Clostridioides difficile/patogenicidade , Infecções por Clostridium/patologia , Colo/patologia , Mucosa Intestinal/patologia , Células-Tronco/patologia , Animais , Proteínas de Bactérias/toxicidade , Toxinas Bacterianas/toxicidade , Células Cultivadas , Clostridioides difficile/metabolismo , Infecções por Clostridium/microbiologia , Colo/citologia , Colo/microbiologia , Modelos Animais de Doenças , Feminino , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Camundongos , Organoides , Cultura Primária de Células , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células-Tronco/microbiologia
4.
Gastroenterology ; 157(4): 1093-1108.e11, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31325428

RESUMO

BACKGROUND & AIMS: Inflammation, injury, and infection up-regulate expression of the tryptophan metabolizing enzyme indoleamine 2,3-dioxygenase 1 (IDO1) in the intestinal epithelium. We studied the effects of cell-specific IDO1 expression in the epithelium at baseline and during intestinal inflammation in mice. METHODS: We generated transgenic mice that overexpress fluorescence-tagged IDO1 in the intestinal epithelium under control of the villin promoter (IDO1-TG). We generated intestinal epithelial spheroids from mice with full-length Ido1 (controls), disruption of Ido1 (knockout mice), and IDO1-TG and analyzed them for stem cell and differentiation markers by real-time polymerase chain reaction, immunoblotting, and immunofluorescence. Some mice were gavaged with enteropathogenic Escherichia coli (E2348/69) to induce infectious ileitis, and ileum contents were quantified by polymerase chain reaction. Separate sets of mice were given dextran sodium sulfate or 2,4,6-trinitrobenzenesulfonic acid to induce colitis; intestinal tissues were analyzed by histology. We utilized published data sets GSE75214 and GDS2642 of RNA expression data from ilea of healthy individuals undergoing screening colonoscopies (controls) and patients with Crohn's disease. RESULTS: Histologic analysis of small intestine tissues from IDO1-TG mice revealed increases in secretory cells. Enteroids derived from IDO1-TG intestine had increased markers of stem, goblet, Paneth, enteroendocrine, and tuft cells, compared with control enteroids, with a concomitant decrease in markers of absorptive cells. IDO1 interacted non-enzymatically with the aryl hydrocarbon receptor to inhibit activation of NOTCH1. Intestinal mucus layers from IDO1-TG mice were 2-fold thicker than mucus layers from control mice, with increased proportions of Akkermansia muciniphila and Mucispirillum schaedleri. Compared to controls, IDO1-TG mice demonstrated an 85% reduction in ileal bacteria (P = .03) when challenged with enteropathogenic E coli, and were protected from immune infiltration, crypt dropout, and ulcers following administration of dextran sodium sulfate or 2,4,6-trinitrobenzenesulfonic acid. In ilea of Crohn's disease patients, increased expression of IDO1 correlated with increased levels of MUC2, LYZ1, and aryl hydrocarbon receptor, but reduced levels of SLC2A5. CONCLUSIONS: In mice, expression of IDO1 in the intestinal epithelial promotes secretory cell differentiation and mucus production; levels of IDO1 are positively correlated with secretory cell markers in ilea of healthy individuals and Crohn's disease patients. We propose that IDO1 contributes to intestinal homeostasis.


Assuntos
Bactérias/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular , Microbioma Gastrointestinal , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Doenças Inflamatórias Intestinais/enzimologia , Doenças Inflamatórias Intestinais/microbiologia , Mucosa Intestinal/enzimologia , Mucosa Intestinal/microbiologia , Receptores de Hidrocarboneto Arílico/metabolismo , Receptores Notch/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Estudos de Casos e Controles , Linhagem Celular , Linhagem da Célula , Modelos Animais de Doenças , Células Epiteliais/enzimologia , Células Epiteliais/microbiologia , Células Epiteliais/patologia , Genótipo , Humanos , Indolamina-Pirrol 2,3,-Dioxigenase/deficiência , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/patologia , Camundongos Knockout , Fenótipo , Receptores de Hidrocarboneto Arílico/genética , Receptores Notch/genética , Via Secretória , Transdução de Sinais , Células-Tronco/enzimologia , Células-Tronco/microbiologia , Células-Tronco/patologia
5.
PLoS Genet ; 13(11): e1007091, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29108021

RESUMO

Cytokine signaling is responsible for coordinating conserved epithelial regeneration and immune responses in the digestive tract. In the Drosophila midgut, Upd3 is a major cytokine, which is induced in enterocytes (EC) and enteroblasts (EB) upon oral infection, and initiates intestinal stem cell (ISC) dependent tissue repair. To date, the genetic network directing upd3 transcription remains largely uncharacterized. Here, we have identified the key infection-responsive enhancers of the upd3 gene and show that distinct enhancers respond to various stresses. Furthermore, through functional genetic screening, bioinformatic analyses and yeast one-hybrid screening, we determined that the transcription factors Scalloped (Sd), Mothers against dpp (Mad), and D-Fos are principal regulators of upd3 expression. Our study demonstrates that upd3 transcription in the gut is regulated by the activation of multiple pathways, including the Hippo, TGF-ß/Dpp, and Src, as well as p38-dependent MAPK pathways. Thus, these essential pathways, which are known to control ISC proliferation cell-autonomously, are also activated in ECs to promote tissue turnover the regulation of upd3 transcription.


Assuntos
Infecções Bacterianas/metabolismo , Drosophila/genética , Drosophila/microbiologia , Transdução de Sinais , Animais , Infecções Bacterianas/genética , Proliferação de Células , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Enterócitos/metabolismo , Feminino , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Intestinos/citologia , Intestinos/microbiologia , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Pectobacterium carotovorum/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Pseudomonas/metabolismo , Células-Tronco/microbiologia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
6.
Infect Immun ; 87(4)2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30642906

RESUMO

The enteric bacterium and intracellular human pathogen Shigella causes hundreds of millions of cases of the diarrheal disease shigellosis per year worldwide. Shigella is acquired by ingestion of contaminated food or water; upon reaching the colon, the bacteria invade colonic epithelial cells, replicate intracellularly, spread to adjacent cells, and provoke an intense inflammatory response. There is no animal model that faithfully recapitulates human disease; thus, cultured cells have been used to model Shigella pathogenesis. However, the use of transformed cells in culture does not provide the same environment to the bacteria as the normal human intestinal epithelium. Recent advances in tissue culture now enable the cultivation of human intestinal enteroids (HIEs), which are derived from human intestinal stem cells, grown ex vivo, and then differentiated into "mini-intestines." Here, we demonstrate that HIEs can be used to model Shigella pathogenesis. We show that Shigella flexneri invades polarized HIE monolayers preferentially via the basolateral surface. After S. flexneri invades HIE monolayers, S. flexneri replicates within HIE cells and forms actin tails. S. flexneri also increases the expression of HIE proinflammatory signals and the amino acid transporter SLC7A5. Finally, we demonstrate that disruption of HIE tight junctions enables S. flexneri invasion via the apical surface.


Assuntos
Disenteria Bacilar/microbiologia , Mucosa Intestinal/microbiologia , Modelos Biológicos , Organoides/microbiologia , Shigella flexneri/fisiologia , Técnicas de Cultura de Células , Disenteria Bacilar/genética , Disenteria Bacilar/metabolismo , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Transportador 1 de Aminoácidos Neutros Grandes/genética , Transportador 1 de Aminoácidos Neutros Grandes/metabolismo , Organoides/crescimento & desenvolvimento , Organoides/metabolismo , Shigella flexneri/patogenicidade , Células-Tronco/citologia , Células-Tronco/microbiologia , Virulência
7.
J Infect Dis ; 217(10): 1667-1671, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29471332

RESUMO

Persistence of Mycobacterium tuberculosis within human bone marrow stem cells has been identified as a potential bacterial niche during latent tuberculosis. Using a murine model of tuberculosis, we show here that bone marrow stem and progenitor cells containing M. tuberculosis propagated tuberculosis when transferred to naive mice, given that both transferred cells and recipient mice were unable to express inducible nitric oxide synthase, which mediates killing of intracellular bacteria via nitric oxide. Our findings suggest that bone marrow stem and progenitor cells containing M. tuberculosis propagate hallmarks of disease if nitric oxide-mediated killing of bacteria is defective.


Assuntos
Células-Tronco Hematopoéticas/metabolismo , Células-Tronco Hematopoéticas/microbiologia , Mycobacterium tuberculosis/patogenicidade , Óxido Nítrico Sintase Tipo II/metabolismo , Células-Tronco/metabolismo , Células-Tronco/microbiologia , Tuberculose/metabolismo , Animais , Modelos Animais de Doenças , Transplante de Células-Tronco Hematopoéticas/métodos , Camundongos , Camundongos Endogâmicos C57BL , Óxido Nítrico/metabolismo , Tuberculose/microbiologia
8.
J Biol Chem ; 292(7): 2586-2600, 2017 02 17.
Artigo em Inglês | MEDLINE | ID: mdl-28053090

RESUMO

Gut microbiota play an important role in regulating the development of the host immune system, metabolic rate, and at times, disease pathogenesis. The factors and mechanisms that mediate interactions between microbiota and the intestinal epithelium are not fully understood. We provide novel evidence that microbiota may control intestinal epithelial stem cell (IESC) proliferation in part through microRNAs (miRNAs). We demonstrate that miRNA profiles differ dramatically across functionally distinct cell types of the mouse jejunal intestinal epithelium and that miRNAs respond to microbiota in a highly cell type-specific manner. Importantly, we also show that miRNAs in IESCs are more prominently regulated by microbiota compared with miRNAs in any other intestinal epithelial cell subtype. We identify miR-375 as one miRNA that is significantly suppressed by the presence of microbiota in IESCs. Using a novel method to knockdown gene and miRNA expression ex vivo enteroids, we demonstrate that we can knock down gene expression in Lgr5+ IESCs. Furthermore, when we knock down miR-375 in IESCs, we observe significantly increased proliferative capacity. Understanding the mechanisms by which microbiota regulate miRNA expression in IESCs and other intestinal epithelial cell subtypes will elucidate a critical molecular network that controls intestinal homeostasis and, given the heightened interest in miRNA-based therapies, may offer novel therapeutic strategies in the treatment of gastrointestinal diseases associated with altered IESC function.


Assuntos
Mucosa Intestinal/metabolismo , Células-Tronco/microbiologia , Transcriptoma , Animais , Feminino , Vida Livre de Germes , Proteínas de Fluorescência Verde/genética , Mucosa Intestinal/citologia , Mucosa Intestinal/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , Fatores de Transcrição SOX9/genética , Células-Tronco/citologia
9.
EMBO J ; 32(23): 3017-28, 2013 Nov 27.
Artigo em Inglês | MEDLINE | ID: mdl-24141879

RESUMO

The resident prokaryotic microbiota of the metazoan gut elicits profound effects on the growth and development of the intestine. However, the molecular mechanisms of symbiotic prokaryotic-eukaryotic cross-talk in the gut are largely unknown. It is increasingly recognized that physiologically generated reactive oxygen species (ROS) function as signalling secondary messengers that influence cellular proliferation and differentiation in a variety of biological systems. Here, we report that commensal bacteria, particularly members of the genus Lactobacillus, can stimulate NADPH oxidase 1 (Nox1)-dependent ROS generation and consequent cellular proliferation in intestinal stem cells upon initial ingestion into the murine or Drosophila intestine. Our data identify and highlight a highly conserved mechanism that symbiotic microorganisms utilize in eukaryotic growth and development. Additionally, the work suggests that specific redox-mediated functions may be assigned to specific bacterial taxa and may contribute to the identification of microbes with probiotic potential.


Assuntos
Proliferação de Células , Drosophila/microbiologia , Intestinos/citologia , Larva/citologia , NADH NADPH Oxirredutases/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/citologia , Animais , Diferenciação Celular , Drosophila/crescimento & desenvolvimento , Drosophila/metabolismo , Histonas/metabolismo , Interações Hospedeiro-Patógeno , Mucosa Intestinal/metabolismo , Intestinos/microbiologia , Lactobacillus/patogenicidade , Larva/metabolismo , Larva/microbiologia , Camundongos , NADPH Oxidase 1 , Oxirredução , Fosforilação , Transdução de Sinais , Células-Tronco/metabolismo , Células-Tronco/microbiologia , Simbiose
10.
Genes Dev ; 23(19): 2333-44, 2009 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-19797770

RESUMO

Gut homeostasis is controlled by both immune and developmental mechanisms, and its disruption can lead to inflammatory disorders or cancerous lesions of the intestine. While the impact of bacteria on the mucosal immune system is beginning to be precisely understood, little is known about the effects of bacteria on gut epithelium renewal. Here, we addressed how both infectious and indigenous bacteria modulate stem cell activity in Drosophila. We show that the increased epithelium renewal observed upon some bacterial infections is a consequence of the oxidative burst, a major defense of the Drosophila gut. Additionally, we provide evidence that the JAK-STAT (Janus kinase-signal transducers and activators of transcription) and JNK (c-Jun NH(2) terminal kinase) pathways are both required for bacteria-induced stem cell proliferation. Similarly, we demonstrate that indigenous gut microbiota activate the same, albeit reduced, program at basal levels. Altered control of gut microbiota in immune-deficient or aged flies correlates with increased epithelium renewal. Finally, we show that epithelium renewal is an essential component of Drosophila defense against oral bacterial infection. Altogether, these results indicate that gut homeostasis is achieved by a complex interregulation of the immune response, gut microbiota, and stem cell activity.


Assuntos
Drosophila melanogaster/citologia , Drosophila melanogaster/microbiologia , Pectobacterium carotovorum/fisiologia , Pseudomonas/fisiologia , Animais , Proliferação de Células , Proteínas de Drosophila/metabolismo , Epitélio/microbiologia , Regulação da Expressão Gênica no Desenvolvimento , Intestinos/citologia , Intestinos/imunologia , Intestinos/microbiologia , Janus Quinases/metabolismo , Explosão Respiratória , Fatores de Transcrição STAT/metabolismo , Células-Tronco/citologia , Células-Tronco/microbiologia , Fatores de Transcrição/metabolismo
11.
Gastroenterology ; 148(7): 1392-404.e21, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25725293

RESUMO

BACKGROUND & AIMS: Helicobacter pylori infection is the main risk factor for gastric cancer. We characterized the interactions of H pylori with gastric epithelial progenitor and stem cells in humans and mice and investigated how these interactions contribute to H pylori-induced pathology. METHODS: We used quantitative confocal microscopy and 3-dimensional reconstruction of entire gastric glands to determine the localizations of H pylori in stomach tissues from humans and infected mice. Using lineage tracing to mark cells derived from leucine-rich repeat-containing G-protein coupled receptor 5-positive (Lgr5(+)) stem cells (Lgr5-eGFP-IRES-CreERT2/Rosa26-TdTomato mice) and in situ hybridization, we analyzed gastric stem cell responses to infection. Isogenic H pylori mutants were used to determine the role of specific virulence factors in stem cell activation and pathology. RESULTS: H pylori grow as distinct bacterial microcolonies deep in the stomach glands and interact directly with gastric progenitor and stem cells in tissues from mice and humans. These gland-associated bacteria activate stem cells, increasing the number of stem cells, accelerating Lgr5(+) stem cell proliferation, and up-regulating expression of stem cell-related genes. Mutant bacteria with defects in chemotaxis that are able to colonize the stomach surface but not the antral glands in mice do not activate stem cells. In addition, bacteria that are unable to inject the contact-dependent virulence factor CagA into the epithelium colonized stomach glands in mice, but did not activate stem cells or produce hyperplasia to the same extent as wild-type H pylori. CONCLUSIONS: H pylori colonize and manipulate the progenitor and stem cell compartments, which alters turnover kinetics and glandular hyperplasia. Bacterial ability to alter the stem cells has important implications for gastrointestinal stem cell biology and H pylori-induced gastric pathology.


Assuntos
Mucosa Gástrica/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/crescimento & desenvolvimento , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/microbiologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Biomarcadores/metabolismo , Proliferação de Células , Modelos Animais de Doenças , Mucosa Gástrica/metabolismo , Genótipo , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/patologia , Helicobacter pylori/genética , Helicobacter pylori/patogenicidade , Interações Hospedeiro-Patógeno , Humanos , Hiperplasia , Cinética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação , Organoides , Fenótipo , Receptores Acoplados a Proteínas G/genética , Células-Tronco/metabolismo , Células-Tronco/patologia , Técnicas de Cultura de Tecidos , Virulência
12.
Gastroenterology ; 148(1): 126-136.e6, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25307862

RESUMO

BACKGROUND & AIMS: We previously established long-term, 3-dimensional culture of organoids from mouse tissues (intestine, stomach, pancreas, and liver) and human intestine and pancreas. Here we describe conditions required for long-term 3-dimensional culture of human gastric stem cells. The technology can be applied to study the epithelial response to infection with Helicobacter pylori. METHODS: We generated organoids from surgical samples of human gastric corpus. Culture conditions were developed based on those for the mouse gastric and human intestinal systems. We used microinjection to infect the organoids with H pylori. Epithelial responses were measured using microarray and quantitative polymerase chain reaction analyses. RESULTS: Human gastric cells were expanded indefinitely in 3-dimensional cultures. We cultured cells from healthy gastric tissues, single-sorted stem cells, or tumor tissues. Organoids maintained many characteristics of their respective tissues based on their histology, expression of markers, and euploidy. Organoids from healthy tissue expressed markers of 4 lineages of the stomach and self-organized into gland and pit domains. They could be directed to specifically express either lineages of the gastric gland, or the gastric pit, by addition of nicotinamide and withdrawal of WNT. Although gastric pit lineages had only marginal reactions to bacterial infection, gastric gland lineages mounted a strong inflammatory response. CONCLUSIONS: We developed a system to culture human gastric organoids. This system can be used to study H pylori infection and other gastric pathologies.


Assuntos
Células Epiteliais/microbiologia , Infecções por Helicobacter/microbiologia , Helicobacter pylori/patogenicidade , Células-Tronco/microbiologia , Estômago/microbiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Linhagem da Célula , Proliferação de Células , Separação Celular , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Mucosa Gástrica/metabolismo , Regulação da Expressão Gênica , Infecções por Helicobacter/imunologia , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Helicobacter pylori/imunologia , Humanos , Masculino , Pessoa de Meia-Idade , Niacinamida/farmacologia , Organoides , Fenótipo , Ploidias , Células-Tronco/efeitos dos fármacos , Células-Tronco/imunologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Estômago/efeitos dos fármacos , Estômago/imunologia , Estômago/patologia , Fatores de Tempo , Proteínas Wnt/metabolismo
13.
Proc Natl Acad Sci U S A ; 110(26): 10788-93, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23744038

RESUMO

Wolbachia are intracellular bacteria that infect invertebrates at pandemic levels, including insect vectors of devastating infectious diseases. Although Wolbachia are providing novel strategies for the control of several human pathogens, the processes underlying Wolbachia's successful propagation within and across species remain elusive. Wolbachia are mainly vertically transmitted; however, there is also evidence of extensive horizontal transmission. Here, we provide several lines of evidence supporting Wolbachia's targeting of ovarian stem cell niches--referred to as "niche tropism"--as a previously overlooked strategy for Wolbachia thriving in nature. Niche tropism is pervasive in Wolbachia infecting the Drosophila genus, and different patterns of niche tropism are evolutionarily conserved. Phylogenetic analysis, confirmed by hybrid introgression and transinfection experiments, demonstrates that bacterial factors are the major determinants of differential patterns of niche tropism. Furthermore, bacterial load is increased in germ-line cells passing through infected niches, supporting previous suggestions of a contribution of Wolbachia from stem-cell niches toward vertical transmission. These results support the role of stem-cell niches as a key component for the spreading of Wolbachia in the Drosophila genus and provide mechanistic insights into this unique tissue tropism.


Assuntos
Drosophila/microbiologia , Ovário/microbiologia , Células-Tronco/microbiologia , Wolbachia/genética , Wolbachia/fisiologia , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/fisiologia , Drosophila/genética , Feminino , Interações Hospedeiro-Patógeno , Masculino , Ovário/citologia , Óvulo/microbiologia , Filogenia , Especificidade da Espécie , Nicho de Células-Tronco , Simbiose , Wolbachia/patogenicidade
14.
Anaerobe ; 37: 25-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26549696

RESUMO

Recent advances in developmental and stem cell biology have resulted in techniques that enable the generation and maintenance of complex epithelium in vitro. While these models have been utilized to study host development and disease, a renewed appreciation of host-microbe interactions has sparked interest in employing these new techniques to study microbes at the epithelial interface. Here we review the current advances in host-microbe interactions that have resulted from experiments using these complex epithelia. Furthermore we highlight aspects of these techniques that warrant further development to facilitate the study of host-microbe interactions.


Assuntos
Mucosa Intestinal/microbiologia , Células-Tronco , Animais , Técnicas Bacteriológicas , Clostridioides difficile/fisiologia , Helicobacter pylori/fisiologia , Humanos , Técnicas In Vitro , Salmonella enterica/fisiologia , Células-Tronco/microbiologia
16.
Eur J Immunol ; 44(8): 2318-30, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24825601

RESUMO

The generation of immune cells from BM precursors is a carefully regulated process. This is essential to limit the potential for oncogenesis and autoimmunity yet protect against infection. How infection modulates this is unclear. Salmonella can colonize systemic sites including the BM and spleen. This resolving infection has multiple IFN-γ-mediated acute and chronic effects on BM progenitors, and during the first week of infection IFN-γ is produced by myeloid, NK, NKT, CD4(+) T cells, and some lineage-negative cells. After infection, the phenotype of BM progenitors rapidly but reversibly alters, with a peak ∼ 30-fold increase in Sca-1(hi) progenitors and a corresponding loss of Sca-1(lo/int) subsets. Most strikingly, the capacity of donor Sca-1(hi) cells to reconstitute an irradiated host is reduced; the longer donor mice are exposed to infection, and Sca-1(hi) c-kit(int) cells have an increased potential to generate B1a-like cells. Thus, Salmonella can have a prolonged influence on BM progenitor functionality not directly related to bacterial persistence. These results reflect changes observed in leucopoiesis during aging and suggest that BM functionality can be modulated by life-long, periodic exposure to infection. Better understanding of this process could offer novel therapeutic opportunities to modulate BM functionality and promote healthy aging.


Assuntos
Células da Medula Óssea/imunologia , Salmonelose Animal/imunologia , Células-Tronco/imunologia , Animais , Antígenos Ly/imunologia , Células da Medula Óssea/microbiologia , Células da Medula Óssea/patologia , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/microbiologia , Homeostase/imunologia , Interferon gama/imunologia , Proteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fenótipo , Salmonella/imunologia , Salmonelose Animal/patologia , Células-Tronco/microbiologia , Células-Tronco/patologia
17.
J Immunol ; 190(4): 1501-9, 2013 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-23325888

RESUMO

CD8(+) T cells play a crucial role in the clearance of intracellular pathogens through the generation of cytotoxic effector cells that eliminate infected cells and long-lived memory cells that provide enhanced protection against reinfection. We have previously shown that the inhibitor of E protein transcription factors, Id2, is necessary for accumulation of effector and memory CD8(+) T cells during infection. In this study, we show that CD8(+) T cells lacking Id2 did not generate a robust terminally differentiated killer cell lectin-like receptor G1 (KLRG1)(hi) effector population, but displayed a cell-surface phenotype and cytokine profile consistent with memory precursors, raising the question as to whether loss of Id2 impairs the differentiation and/or survival of effector memory cells. We found that deletion of Bim rescued Id2-deficient CD8(+) cell survival during infection. However, the dramatic reduction in KLRG1(hi) cells caused by loss of Id2 remained in the absence of Bim, such that Id2/Bim double-deficient cells form an exclusively KLRG1(lo)CD127(hi) memory precursor population. Thus, we describe a role for Id2 in both the survival and differentiation of normal CD8(+) effector and memory populations.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteína 2 Inibidora de Diferenciação/fisiologia , Receptores Imunológicos/biossíntese , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/microbiologia , Linfócitos T CD8-Positivos/virologia , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Citocinas/biossíntese , Memória Imunológica/genética , Imunofenotipagem , Proteína 2 Inibidora de Diferenciação/deficiência , Proteína 2 Inibidora de Diferenciação/genética , Subunidade alfa de Receptor de Interleucina-7/biossíntese , Lectinas Tipo C , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Células-Tronco/imunologia , Células-Tronco/microbiologia , Células-Tronco/virologia , Proteína bcl-X/deficiência , Proteína bcl-X/genética
19.
Clin Oral Investig ; 19(8): 2059-66, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25689981

RESUMO

OBJECTIVES: To investigate the effect of various dilutions of antibiotic medicaments used in endodontic regeneration on the survival of human dental pulp stem cells (DPSCs) and to determine their antibacterial effect against established Enterococcus faecalis biofilm. MATERIALS AND METHODS: The cytotoxic and antibacterial effects of different triple (TAP) and double antibiotic paste (DAP) dilutions (0.125, 0.25, 0.5, 1, and 10 mg/ml) were tested against Enterococcus faecalis established biofilm and DPSC. Established bacterial biofilm were exposed to antibiotic dilutions for 3 days. Then, biofilms were collected, spiral plated, and the numbers of bacterial colony forming units (CFU/ml) were determined. For the cytotoxic effect, lactate dehydrogenase activity assays (LDH) and cell viability assays (WST-1) were used to measure the percentage of DPSC cytotoxicity after 3-day treatment with the same antibiotic dilutions. A general linear mixed model was used for statistical analyses (α = 0.05). RESULTS: All antibiotic dilutions significantly decreased the bacterial CFU/ml. For WST-1 assays, all antibiotic dilutions except 0.125 mg/ml significantly reduced the viability of DPSC. For LDH assays, the three lowest tested concentrations of DAP (0.5, 0.25, 0.125 mg/ml) and the two lowest concentrations of TAP (0.25 and 0.125 mg/ml) were non-toxic to DPSC. CONCLUSIONS: All tested dilutions had an antibacterial effect against E. faecalis. However, 0.125 mg/ml of DAP and TAP showed a significant antibacterial effect with no cytotoxic effects on DPSCs. CLINICAL RELEVANCE: Using appropriate antibiotic concentrations of intracanal medicament during endodontic regeneration procedures is critical to disinfect root canal and decrease the adverse effects on stem cells.


Assuntos
Antibacterianos/farmacologia , Biofilmes/crescimento & desenvolvimento , Polpa Dentária/microbiologia , Enterococcus faecalis/fisiologia , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Células-Tronco/microbiologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Polpa Dentária/metabolismo , Polpa Dentária/patologia , Infecções por Bactérias Gram-Positivas/metabolismo , Infecções por Bactérias Gram-Positivas/microbiologia , Infecções por Bactérias Gram-Positivas/patologia , Humanos , Pomadas/farmacologia , Células-Tronco/metabolismo , Células-Tronco/patologia , Fatores de Tempo
20.
J Agric Food Chem ; 72(18): 10366-10375, 2024 May 08.
Artigo em Inglês | MEDLINE | ID: mdl-38651967

RESUMO

Intestinal stem cells (ISCs) sustain epithelial renewal by dynamically altering behaviors of proliferation and differentiation in response to various nutrition and stress inputs. However, how ISCs integrate bioactive substance morin cues to protect against heat-stable enterotoxin b (STb) produced by Escherichia coli remains an uncertain question with implications for treating bacterial diarrhea. Our recent work showed that oral mulberry leaf-derived morin improved the growth performance in STb-challenged mice. Furthermore, morin supplementation reinstated the impaired small-intestinal epithelial structure and barrier function by stimulating ISC proliferation and differentiation as well as supporting intestinal organoid expansion ex vivo. Importantly, the Wnt/ß-catenin pathway, an ISC fate commitment signal, was reactivated by morin to restore the jejunal crypt-villus architecture in response to STb stimulation. Mechanically, the extracellular morin-initiated ß-catenin axis is dependent or partially dependent on the Wnt membrane receptor Frizzled7 (FZD7). Our data reveal an unexpected role of leaf-derived morin, which represents molecular signaling targeting the FZD7 platform instrumental for controlling ISC regeneration upon STb injury.


Assuntos
Antioxidantes , Toxinas Bacterianas , Enterotoxinas , Infecções por Escherichia coli , Proteínas de Escherichia coli , Jejuno , Morus , Extratos Vegetais , Camundongos , Morus/química , Folhas de Planta/química , Via de Sinalização Wnt , Células-Tronco/efeitos dos fármacos , Células-Tronco/microbiologia , Células-Tronco/patologia , Proteínas de Escherichia coli/metabolismo , Técnicas In Vitro , Extratos Vegetais/farmacologia , Jejuno/efeitos dos fármacos , Jejuno/metabolismo , Jejuno/microbiologia , Jejuno/patologia , Regeneração , Toxinas Bacterianas/isolamento & purificação , Enterotoxinas/isolamento & purificação , Infecções por Escherichia coli/tratamento farmacológico , Antioxidantes/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA