Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Cell Mol Gastroenterol Hepatol ; 9(3): 507-526, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31778828

RESUMO

BACKGROUND & AIMS: The mucus layer in the human colon protects against commensal bacteria and pathogens, and defects in its unique bilayered structure contribute to intestinal disorders, such as ulcerative colitis. However, our understanding of colon physiology is limited by the lack of in vitro models that replicate human colonic mucus layer structure and function. Here, we investigated if combining organ-on-a-chip and organoid technologies can be leveraged to develop a human-relevant in vitro model of colon mucus physiology. METHODS: A human colon-on-a-chip (Colon Chip) microfluidic device lined by primary patient-derived colonic epithelial cells was used to recapitulate mucus bilayer formation, and to visualize mucus accumulation in living cultures noninvasively. RESULTS: The Colon Chip supports spontaneous goblet cell differentiation and accumulation of a mucus bilayer with impenetrable and penetrable layers, and a thickness similar to that observed in the human colon, while maintaining a subpopulation of proliferative epithelial cells. Live imaging of the mucus layer formation on-chip showed that stimulation of the colonic epithelium with prostaglandin E2, which is increased during inflammation, causes rapid mucus volume expansion via an Na-K-Cl cotransporter 1 ion channel-dependent increase in its hydration state, but no increase in de novo mucus secretion. CONCLUSIONS: This study shows the production of colonic mucus with a physiologically relevant bilayer structure in vitro, which can be analyzed in real time noninvasively. The Colon Chip may offer a new preclinical tool to analyze the role of mucus in human intestinal homeostasis as well as diseases, such as ulcerative colitis and cancer.


Assuntos
Colo/metabolismo , Mucosa Intestinal/metabolismo , Dispositivos Lab-On-A-Chip , Muco/metabolismo , Células Cultivadas , Dinoprostona/metabolismo , Células Caliciformes/fisiologia , Humanos , Organoides , Cultura Primária de Células/métodos , Membro 1 da Família 12 de Carreador de Soluto/metabolismo
2.
Microbiome ; 7(1): 43, 2019 03 20.
Artigo em Inglês | MEDLINE | ID: mdl-30890187

RESUMO

BACKGROUND: Species-specific differences in tolerance to infection are exemplified by the high susceptibility of humans to enterohemorrhagic Escherichia coli (EHEC) infection, whereas mice are relatively resistant to this pathogen. This intrinsic species-specific difference in EHEC infection limits the translation of murine research to human. Furthermore, studying the mechanisms underlying this differential susceptibility is a difficult problem due to complex in vivo interactions between the host, pathogen, and disparate commensal microbial communities. RESULTS: We utilize organ-on-a-chip (Organ Chip) microfluidic culture technology to model damage of the human colonic epithelium induced by EHEC infection, and show that epithelial injury is greater when exposed to metabolites derived from the human gut microbiome compared to mouse. Using a multi-omics approach, we discovered four human microbiome metabolites-4-methyl benzoic acid, 3,4-dimethylbenzoic acid, hexanoic acid, and heptanoic acid-that are sufficient to mediate this effect. The active human microbiome metabolites preferentially induce expression of flagellin, a bacterial protein associated with motility of EHEC and increased epithelial injury. Thus, the decreased tolerance to infection observed in humans versus other species may be due in part to the presence of compounds produced by the human intestinal microbiome that actively promote bacterial pathogenicity. CONCLUSION: Organ-on-chip technology allowed the identification of specific human microbiome metabolites modulating EHEC pathogenesis. These identified metabolites are sufficient to increase susceptibility to EHEC in our human Colon Chip model and they contribute to species-specific tolerance. This work suggests that higher concentrations of these metabolites could be the reason for higher susceptibility to EHEC infection in certain human populations, such as children. Furthermore, this research lays the foundation for therapeutic-modulation of microbe products in order to prevent and treat human bacterial infection.


Assuntos
Bactérias/metabolismo , Escherichia coli Êntero-Hemorrágica/patogenicidade , Infecções por Escherichia coli/patologia , Intestinos/citologia , Técnicas de Cultura de Órgãos/métodos , Animais , Benzoatos/farmacologia , Caproatos/farmacologia , Células Cultivadas , Escherichia coli Êntero-Hemorrágica/metabolismo , Infecções por Escherichia coli/microbiologia , Feminino , Microbioma Gastrointestinal , Ácidos Heptanoicos/farmacologia , Humanos , Intestinos/microbiologia , Masculino , Camundongos , Procedimentos Analíticos em Microchip , Especificidade da Espécie
4.
Sci Rep ; 8(1): 2871, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29440725

RESUMO

Here we describe a method for fabricating a primary human Small Intestine-on-a-Chip (Intestine Chip) containing epithelial cells isolated from healthy regions of intestinal biopsies. The primary epithelial cells are expanded as 3D organoids, dissociated, and cultured on a porous membrane within a microfluidic device with human intestinal microvascular endothelium cultured in a parallel microchannel under flow and cyclic deformation. In the Intestine Chip, the epithelium forms villi-like projections lined by polarized epithelial cells that undergo multi-lineage differentiation similar to that of intestinal organoids, however, these cells expose their apical surfaces to an open lumen and interface with endothelium. Transcriptomic analysis also indicates that the Intestine Chip more closely mimics whole human duodenum in vivo when compared to the duodenal organoids used to create the chips. Because fluids flowing through the lumen of the Intestine Chip can be collected continuously, sequential analysis of fluid samples can be used to quantify nutrient digestion, mucus secretion and establishment of intestinal barrier function over a period of multiple days in vitro. The Intestine Chip therefore may be useful as a research tool for applications where normal intestinal function is crucial, including studies of metabolism, nutrition, infection, and drug pharmacokinetics, as well as personalized medicine.


Assuntos
Intestino Delgado/citologia , Dispositivos Lab-On-A-Chip , Organoides/citologia , Biópsia , Proliferação de Células , Células Epiteliais/citologia , Humanos
5.
Stem Cell Reports ; 10(1): 17-26, 2018 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-29276155

RESUMO

The intestinal epithelium serves as an essential barrier to the outside world and is maintained by functionally distinct populations of rapidly cycling intestinal stem cells (CBC ISCs) and slowly cycling, reserve ISCs (r-ISCs). Because disruptions in the epithelial barrier can result from pathological activation of the immune system, we sought to investigate the impact of inflammation on ISC behavior during the regenerative response. In a murine model of αCD3 antibody-induced small-intestinal inflammation, r-ISCs proved highly resistant to injury, while CBC ISCs underwent apoptosis. Moreover, r-ISCs were induced to proliferate and functionally contribute to intestinal regeneration. Further analysis revealed that the inflammatory cytokines interferon gamma and tumor necrosis factor alpha led to r-ISC activation in enteroid culture, which could be blocked by the JAK/STAT inhibitor, tofacitinib. These results highlight an important role for r-ISCs in response to acute intestinal inflammation and show that JAK/STAT-1 signaling is required for the r-ISC regenerative response.


Assuntos
Enterite/metabolismo , Mucosa Intestinal/fisiologia , Intestino Delgado/metabolismo , Janus Quinases/metabolismo , Regeneração , Fator de Transcrição STAT1/metabolismo , Transdução de Sinais , Células-Tronco/metabolismo , Doença Aguda , Animais , Apoptose/efeitos dos fármacos , Citocinas/metabolismo , Enterite/induzido quimicamente , Enterite/patologia , Inflamação/induzido quimicamente , Inflamação/metabolismo , Inflamação/patologia , Mucosa Intestinal/patologia , Intestino Delgado/patologia , Janus Quinases/antagonistas & inibidores , Camundongos , Camundongos Transgênicos , Piperidinas/farmacologia , Pirimidinas/farmacologia , Pirróis/farmacologia , Fator de Transcrição STAT1/antagonistas & inibidores , Células-Tronco/patologia
6.
Dev Dyn ; 245(7): 718-26, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27153394

RESUMO

The intestine's ability to recover from catastrophic injury requires quiescent intestinal stem cells (q-ISCs). While rapidly cycling (Lgr5+) crypt base columnar (CBC) ISCs normally maintain the intestine, they are highly sensitive to pathological injuries (irradiation, inflammation) and must be restored by q-ISCs to sustain intestinal homeostasis. Despite clear relevance to human health, virtually nothing is known regarding the factors that regulate q-ISCs. A comprehensive understanding of these mechanisms would likely lead to targeted new therapies with profound therapeutic implications for patients with gastrointestinal conditions. We briefly review the current state of the literature, highlighting homeostatic mechanisms important for q-ISC regulation, listing key questions in the field, and offer strategies to address them. Developmental Dynamics 245:718-726, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Mucosa Intestinal/metabolismo , Intestinos/citologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Homeostase/genética , Homeostase/fisiologia , Humanos , Modelos Biológicos , Transdução de Sinais , Via de Sinalização Wnt/genética , Via de Sinalização Wnt/fisiologia
7.
Cell Stem Cell ; 18(3): 410-21, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-26908146

RESUMO

The gastrointestinal (GI) epithelium is a highly regenerative tissue with the potential to provide a renewable source of insulin(+) cells after undergoing cellular reprogramming. Here, we show that cells of the antral stomach have a previously unappreciated propensity for conversion into functional insulin-secreting cells. Native antral endocrine cells share a surprising degree of transcriptional similarity with pancreatic ß cells, and expression of ß cell reprogramming factors in vivo converts antral cells efficiently into insulin(+) cells with close molecular and functional similarity to ß cells. Induced GI insulin(+) cells can suppress hyperglycemia in a diabetic mouse model for at least 6 months and regenerate rapidly after ablation. Reprogramming of antral stomach cells assembled into bioengineered mini-organs in vitro yielded transplantable units that also suppressed hyperglycemia in diabetic mice, highlighting the potential for development of engineered stomach tissues as a renewable source of functional ß cells for glycemic control.


Assuntos
Técnicas de Reprogramação Celular , Reprogramação Celular , Mucosa Gástrica/metabolismo , Células Secretoras de Insulina/metabolismo , Animais , Mucosa Gástrica/citologia , Mucosa Gástrica/transplante , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/transplante , Camundongos
8.
J Physiol ; 594(17): 4805-13, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-26670741

RESUMO

Long-lived and self-renewing adult stem cells (SCs) are essential for homeostasis in a wide range of tissues and can include both rapidly cycling and quiescent (q)SC populations. Rapidly cycling SCs function principally during normal tissue maintenance and are highly sensitive to stress, whereas qSCs exit from their quiescent state in response to homeostatic imbalance and regenerative pressure. The regulatory mechanisms underlying the quiescent state include factors essential for cell cycle control, stress response and survival pathways, developmental signalling pathways, and post-transcriptional modulation. Here, we review these regulatory mechanisms citing observations from the intestine and other self-renewing tissues.


Assuntos
Células-Tronco Adultas/fisiologia , Intestinos/citologia , Animais , Autofagia , Proteínas Inibidoras de Quinase Dependente de Ciclina/fisiologia , Proteínas de Ligação a DNA/fisiologia , PTEN Fosfo-Hidrolase/fisiologia , Proteínas Quinases/fisiologia
9.
Cell Rep ; 13(11): 2403-2411, 2015 Dec 22.
Artigo em Inglês | MEDLINE | ID: mdl-26686631

RESUMO

The cellular and molecular mechanisms underlying adaptive changes to physiological stress within the intestinal epithelium remain poorly understood. Here, we show that PTEN, a negative regulator of the PI3K→AKT→mTORC1-signaling pathway, is an important regulator of dormant intestinal stem cells (d-ISCs). Acute nutrient deprivation leads to transient PTEN phosphorylation within d-ISCs and a corresponding increase in their number. This release of PTEN inhibition renders d-ISCs functionally poised to contribute to the regenerative response during re-feeding via cell-autonomous activation of the PI3K→AKT→mTORC1 pathway. Consistent with its role in mediating cell survival, PTEN is required for d-ISC maintenance at baseline, and intestines lacking PTEN have diminished regenerative capacity after irradiation. Our results highlight a PTEN-dependent mechanism for d-ISC maintenance and further demonstrate the role of d-ISCs in the intestinal response to stress.


Assuntos
Intestinos/citologia , Estado Nutricional , PTEN Fosfo-Hidrolase/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Proliferação de Células , Feminino , Genes Reporter , Intestinos/patologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Complexos Multiproteicos/metabolismo , PTEN Fosfo-Hidrolase/genética , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Telomerase/genética , Telomerase/metabolismo
10.
Proc Natl Acad Sci U S A ; 108(1): 179-84, 2011 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-21173232

RESUMO

The intestinal epithelium is maintained by a population of rapidly cycling (Lgr5(+)) intestinal stem cells (ISCs). It has been postulated, however, that slowly cycling ISCs must also be present in the intestine to protect the genome from accumulating deleterious mutations and to allow for a response to tissue injury. Here, we identify a subpopulation of slowly cycling ISCs marked by mouse telomerase reverse transcriptase (mTert) expression that can give rise to Lgr5(+) cells. mTert-expressing cells distribute in a pattern along the crypt-villus axis similar to long-term label-retaining cells (LRCs) and are resistant to tissue injury. Lineage-tracing studies demonstrate that mTert(+) cells give rise to all differentiated intestinal cell types, persist long term, and contribute to the regenerative response following injury. Consistent with other highly regenerative tissues, our results demonstrate that a slowly cycling stem cell population exists within the intestine.


Assuntos
Mucosa Intestinal/citologia , Células-Tronco Multipotentes/metabolismo , Telomerase/metabolismo , Animais , Linhagem da Célula/fisiologia , Citometria de Fluxo , Proteínas de Fluorescência Verde/metabolismo , Imuno-Histoquímica , Camundongos , Microscopia de Fluorescência , Células-Tronco Multipotentes/citologia , Receptores Acoplados a Proteínas G/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
11.
Prog Mol Biol Transl Sci ; 96: 207-29, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-21075346

RESUMO

Regulation of gene expression within the intestinal epithelium is complex and controlled by various signaling pathways that regulate the balance between proliferation and differentiation. Proliferation is required both to grow and to replace cells lost through apoptosis and attrition, yet in all but a few cells, differentiation must take place to prevent uncontrolled growth (cancer) and to provide essential functions. In this chapter, we review the major signaling pathways underlying regulation of gene expression within the intestinal epithelium, based primarily on data from mouse models, as well as specific morphogens and transcription factor families that have a major role in regulating intestinal gene expression, including the Hedgehog family, Forkhead Box (FOX) factors, Homeobox (HOX) genes, ParaHox genes, GATA transcription factors, canonical Wnt/ß-catenin signaling, EPH/Ephrins, Sox9, BMP signaling, PTEN/PI3K, LKB1, K-RAS, Notch pathway, HNF, and MATH1. We also briefly highlight important emerging areas of gene regulation, including microRNA (miRNA) and epigenetic regulation.


Assuntos
Regulação da Expressão Gênica , Mucosa Intestinal/metabolismo , Animais , Humanos , Transdução de Sinais/genética , Fatores de Transcrição/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA