Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell Mol Gastroenterol Hepatol ; 12(4): 1479-1502, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34242819

RESUMO

BACKGROUND & AIMS: CD4+ T cells are regulated by activating and inhibitory cues, and dysregulation of these proper regulatory inputs predisposes these cells to aberrant inflammation and exacerbation of disease. We investigated the role of the inhibitory receptor paired immunoglobulin-like receptor B (PIR-B) in the regulation of the CD4+ T-cell inflammatory response and exacerbation of the colitic phenotype. METHODS: We used Il10-/- spontaneous and CD4+CD45RBhi T-cell transfer models of colitis with PIR-B-deficient (Pirb-/-) mice. Flow cytometry, Western blot, and RNA sequencing analysis was performed on wild-type and Pirb-/- CD4+ T cells. In silico analyses were performed on RNA sequencing data set of ileal biopsy samples from pediatric CD and non-inflammatory bowel disease patients and sorted human memory CD4+ T cells. RESULTS: We identified PIR-B expression on memory CD4+ interleukin (IL)17a+ cells. We show that PIR-B regulates CD4+ T-helper 17 cell (Th17)-dependent chronic intestinal inflammatory responses and the development of colitis. Mechanistically, we show that the PIR-B- Src-homology region 2 domain-containing phosphatase-1/2 axis tempers mammalian target of rapamycin complex 1 signaling and mammalian target of rapamycin complex 1-dependent caspase-3/7 apoptosis, resulting in CD4+ IL17a+ cell survival. In silico analyses showed enrichment of transcriptional signatures for Th17 cells (RORC, RORA, and IL17A) and tissue resident memory (HOBIT, IL7R, and BLIMP1) networks in PIR-B+ murine CD4+ T cells and human CD4+ T cells that express the human homologue leukocyte immunoglobulin-like receptor subfamily B member 3 (LILRB3). High levels of LILRB3 expression were associated strongly with mucosal injury and a proinflammatory Th17 signature, and this signature was restricted to a treatment-naïve, severe pediatric CD population. CONCLUSIONS: Our findings show an intrinsic role for PIR-B/LILRB3 in the regulation of CD4+ IL17a+ T-cell pathogenic memory responses.


Assuntos
Regulação da Expressão Gênica , Imunomodulação , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Receptores Imunológicos/genética , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Animais , Biomarcadores , Sobrevivência Celular/genética , Sobrevivência Celular/imunologia , Colite/etiologia , Colite/metabolismo , Colite/patologia , Modelos Animais de Doenças , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Imuno-Histoquímica , Memória Imunológica , Imunofenotipagem , Doenças Inflamatórias Intestinais/etiologia , Doenças Inflamatórias Intestinais/metabolismo , Doenças Inflamatórias Intestinais/patologia , Interleucina-10/genética , Interleucina-10/metabolismo , Interleucina-17/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Receptores Imunológicos/metabolismo , Transdução de Sinais
2.
Front Immunol ; 12: 636198, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33841417

RESUMO

Food allergy is an emerging epidemic, and the underlying mechanisms are not well defined partly due to the lack of robust adjuvant free experimental models of dietary antigen sensitization. As housing mice at thermoneutrality (Tn) - the temperature of metabolic homeostasis (26-30°C) - has been shown to improve modeling various human diseases involved in inflammation, we tested the impact of Tn housing on an experimental model of food sensitization. Here we demonstrate that WT BALB/c mice housed under standard temperature (18-20°C, Ts) conditions translocated the luminal antigens in the small intestine (SI) across the epithelium via goblet cell antigen passages (GAPs). In contrast, food allergy sensitive Il4raF709 mice housed under standard temperature conditions translocated the luminal antigens in the SI across the epithelium via secretory antigen passages (SAPs). Activation of SI antigen passages and oral challenge of Il4raF709 mice with egg allergens at standard temperature predisposed Il4raF709 mice to develop an anaphylactic reaction. Housing Il4raF709 mice at Tn altered systemic type 2 cytokine, IL-4, and the landscape of SI antigen passage patterning (villus and crypt involvement). Activation of SI antigen passages and oral challenge of Il4raF709 mice with egg antigen under Tn conditions led to the robust induction of egg-specific IgE and development of food-induced mast cell activation and hypovolemic shock. Similarly, Tn housing of WT BALB/c mice altered the cellular patterning of SI antigen passage (GAPs to SAPs). Activation of SI antigen passages and the oral challenge of WT BALB/c mice with egg antigen led to systemic reactivity to egg and mast cell activation. Together these data demonstrate that Tn housing alters antigen passage cellular patterning and landscape, and concurrent oral exposure of egg antigens and SAP activation is sufficient to induce oral antigen sensitization.


Assuntos
Alérgenos/metabolismo , Anafilaxia/metabolismo , Hipersensibilidade a Ovo/metabolismo , Proteínas do Ovo/metabolismo , Abrigo para Animais , Intestino Delgado/metabolismo , Temperatura , Administração Oral , Alérgenos/administração & dosagem , Alérgenos/imunologia , Anafilaxia/imunologia , Anafilaxia/microbiologia , Animais , Modelos Animais de Doenças , Hipersensibilidade a Ovo/imunologia , Hipersensibilidade a Ovo/microbiologia , Proteínas do Ovo/administração & dosagem , Proteínas do Ovo/imunologia , Microbioma Gastrointestinal , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Células Caliciformes/microbiologia , Intestino Delgado/imunologia , Intestino Delgado/microbiologia , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Knockout , Permeabilidade , Receptores de Superfície Celular/genética , Receptores de Superfície Celular/metabolismo
3.
Mucosal Immunol ; 14(1): 135-143, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-32576925

RESUMO

Food-triggered anaphylaxis can encompass a variety of systemic and intestinal symptoms. Murine-based and clinical studies have revealed a role for histamine and H1R and H2R-pathway in the systemic response; however, the molecular processes that regulate the gastrointestinal (GI) response are not as well defined. In the present study, by utilizing an IgE-mast cell (MC)-dependent experimental model of oral antigen-induced anaphylaxis, we define the intestinal epithelial response during a food-induced anaphylactic reaction. We show that oral allergen-challenge stimulates a rapid dysregulation of intestinal epithelial transcellular and paracellular transport that was associated with the development of secretory diarrhea. Allergen-challenge induced (1) a rapid intestinal epithelial Cftr-dependent Cl- secretory response and (2) paracellular macromolecular leak that was associated with modification in epithelial intercellular junction proteins claudin-1, 2, 3 and 5, E-cadherin and desmosomal cadherins. OVA-induced Cftr-dependent Cl- secretion and junctional protein degradation was rapid occurring and was sustained for 72 h following allergen-challenge. Blockade of both the proteolytic activity and Cl- secretory response was required to alleviate intestinal symptoms of food-induced anaphylaxis. Collectively, these data suggest that the GI symptom of food-induced anaphylactic reaction, secretory diarrhea, is a consequence of CFTR-dependent Cl- secretion and proteolytic activity.


Assuntos
Anafilaxia/etiologia , Anafilaxia/metabolismo , Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Hipersensibilidade Alimentar/etiologia , Hipersensibilidade Alimentar/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Alérgenos/imunologia , Anafilaxia/patologia , Animais , Biomarcadores , Modelos Animais de Doenças , Suscetibilidade a Doenças , Hipersensibilidade Alimentar/patologia , Imunoglobulina E/imunologia , Transporte de Íons , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos
4.
J Allergy Clin Immunol ; 145(1): 239-254.e2, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31647967

RESUMO

BACKGROUND: The pathology of eosinophilic esophagitis (EoE) is characterized by eosinophil-rich inflammation, basal zone hyperplasia (BZH), and dilated intercellular spaces, and the underlying processes that drive the pathologic manifestations of the disease remain largely unexplored. OBJECTIVE: We sought to investigate the involvement of the calcium-activated chloride channel anoctamin 1 (ANO1) in esophageal proliferation and the histopathologic features of EoE. METHODS: We examined mRNA and protein expression of ANO1 in esophageal biopsy samples from patients with EoE and in mice with EoE. We performed molecular and cellular analyses and ion transport assays on an in vitro esophageal epithelial 3-dimensional model system (EPC2-ALI) and murine models of EoE to define the relationship between expression and function of ANO1 and esophageal epithelial proliferation in patients with EoE. RESULTS: We observed increased ANO1 expression in esophageal biopsy samples from patients with EoE and in mice with EoE. ANO1 was expressed within the esophageal basal zone, and expression correlated positively with disease severity (eosinophils/high-power field) and BZH. Using an in vitro esophageal epithelial 3-dimensional model system revealed that ANO1 undergoes chromatin modification and rapid upregulation of expression after IL-13 stimulation, that ANO1 is the primary apical IL-13-induced Cl- transport mechanism within the esophageal epithelium, and that loss of ANO1-dependent Cl- transport abrogated esophageal epithelial proliferation. Mechanistically, ANO1-dependent regulation of basal cell proliferation was associated with modulation of TP63 expression and phosphorylated cyclin-dependent kinase 2 levels. CONCLUSIONS: These data identify a functional role for ANO1 in esophageal cell proliferation and BZH in patients with EoE and provide a rationale for pharmacologic intervention of ANO1 function in patients with EoE.


Assuntos
Anoctamina-1/imunologia , Esofagite Eosinofílica/imunologia , Células Epiteliais/imunologia , Esôfago/imunologia , Regulação da Expressão Gênica , Proteínas de Neoplasias/imunologia , Animais , Linhagem Celular , Modelos Animais de Doenças , Esofagite Eosinofílica/patologia , Células Epiteliais/patologia , Esôfago/patologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C
5.
J Allergy Clin Immunol ; 144(4): 1058-1073.e3, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31175877

RESUMO

BACKGROUND: Food-induced anaphylaxis (FIA) is an IgE-dependent immune response that can affect multiple organs and lead to life-threatening complications. The processes by which food allergens cross the mucosal surface and are delivered to the subepithelial immune compartment to promote the clinical manifestations associated with food-triggered anaphylaxis are largely unexplored. OBJECTIVE: We sought to define the processes involved in the translocation of food allergens across the mucosal epithelial surface to the subepithelial immune compartment in FIA. METHODS: Two-photon confocal and immunofluorescence microscopy was used to visualize and trace food allergen passage in a murine model of FIA. A human colon cancer cell line, RNA silencing, and pharmacologic approaches were used to identify the molecular regulation of intestinal epithelial allergen uptake and translocation. Human intestinal organoid transplants were used to demonstrate the conservation of these molecular processes in human tissues. RESULTS: Food allergens are sampled by using small intestine (SI) epithelial secretory cells (termed secretory antigen passages [SAPs]) that are localized to the SI villous and crypt region. SAPs channel food allergens to lamina propria mucosal mast cells through an IL-13-CD38-cyclic adenosine diphosphate ribose (cADPR)-dependent process. Blockade of IL-13-induced CD38/cADPR-dependent SAP antigen passaging in mice inhibited induction of clinical manifestations of FIA. IL-13-CD38-cADPR-dependent SAP sampling of food allergens was conserved in human intestinal organoids. CONCLUSION: We identify that SAPs are a mechanism by which food allergens are channeled across the SI epithelium mediated by the IL-13/CD38/cADPR pathway, regulate the onset of FIA reactions, and are conserved in human intestine.


Assuntos
Alérgenos/imunologia , Anafilaxia/imunologia , Hipersensibilidade Alimentar/imunologia , Interleucina-13/imunologia , Mucosa Intestinal/imunologia , Alérgenos/metabolismo , Anafilaxia/metabolismo , Animais , Hipersensibilidade Alimentar/metabolismo , Humanos , Imunoglobulina E/imunologia , Interleucina-13/metabolismo , Mucosa Intestinal/metabolismo , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos SCID
6.
J Allergy Clin Immunol ; 142(6): 1843-1855, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29729938

RESUMO

BACKGROUND: Eosinophilic esophagitis (EoE) is characterized by histopathologic modifications of esophageal tissue, including eosinophil-rich inflammation, basal zone hyperplasia, and dilated intercellular spaces (DIS). The underlying molecular processes that drive the histopathologic features of EoE remain largely unexplored. OBJECTIVE: We sought to investigate the involvement of solute carrier family 9, subfamily A, member 3 (SLC9A3) in esophageal epithelial intracellular pH (pHi) and DIS formation and the histopathologic features of EoE. METHODS: We examined expression of esophageal epithelial gene networks associated with regulation of pHi in the EoE transcriptome of primary esophageal epithelial cells and an in vitro esophageal epithelial 3-dimensional model system (EPC2-ALI). Molecular and cellular analyses and ion transport assays were used to evaluate the expression and function of SLC9A3. RESULTS: We identified altered expression of gene networks associated with regulation of pHi and acid-protective mechanisms in esophageal biopsy specimens from pediatric patients with EoE (healthy subjects, n = 6; patients with EoE, n = 10). The most dysregulated gene central to regulating pHi was SLC9A3. SLC9A3 expression was increased within the basal layer of esophageal biopsy specimens from patients with EoE, and expression positively correlated with disease severity (eosinophils/high-power field) and DIS (healthy subjects, n = 10; patients with EoE, n = 10). Analyses of esophageal epithelial cells revealed IL-13-induced, signal transducer and activator of transcription 6-dependent SLC9A3 expression and Na+-dependent proton secretion and that SLC9A3 activity correlated positively with DIS formation. Finally, we showed that IL-13-mediated, Na+-dependent proton secretion was the primary intracellular acid-protective mechanism within the esophageal epithelium and that blockade of SLC9A3 transport abrogated IL-13-induced DIS formation. CONCLUSIONS: SLC9A3 plays a functional role in DIS formation, and pharmacologic interventions targeting SLC9A3 function may suppress the histopathologic manifestations in patients with EoE.


Assuntos
Esofagite Eosinofílica/metabolismo , Células Epiteliais/química , Espaço Extracelular , Trocador 3 de Sódio-Hidrogênio/metabolismo , Linhagem Celular , Esofagite Eosinofílica/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Esôfago/patologia , Guanidinas/farmacologia , Humanos , Concentração de Íons de Hidrogênio , Interleucina-13/farmacologia , Metacrilatos/farmacologia , Trocador 3 de Sódio-Hidrogênio/antagonistas & inibidores
7.
J Allergy Clin Immunol ; 142(4): 1159-1172.e5, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29157947

RESUMO

BACKGROUND: Severe IgE-mediated, food-induced anaphylactic reactions are characterized by pulmonary venous vasodilatation and fluid extravasation, which are thought to lead to the life-threatening anaphylactic phenotype. The underlying immunologic and cellular processes involved in driving fluid extravasation and the severe anaphylactic phenotype are not fully elucidated. OBJECTIVE: We sought to define the interaction and requirement of IL-4 and vascular endothelial (VE) IL-4 receptor α chain (IL-4Rα) signaling in histamine-abelson murine leukemia viral oncogene homology 1 (ABL1)-mediated VE dysfunction and fluid extravasation in the severity of IgE-mediated anaphylactic reactions in mice. METHODS: Mice deficient in VE IL-4Rα and models of passive and active oral antigen- and IgE-induced anaphylaxis were used to define the requirements of the VE IL-4Rα and ABL1 pathway in severe anaphylactic reactions. The human VE cell line (EA.hy926 cells) and pharmacologic (imatinib) and genetic (short hairpin RNA knockdown of IL4RA and ABL1) approaches were used to define the requirement of this pathway in VE barrier dysfunction. RESULTS: IL-4 exacerbation of histamine-induced hypovolemic shock in mice was dependent on VE expression of IL-4Rα. IL-4- and histamine-induced ABL1 activation in human VE cells and VE barrier dysfunction was ABL1-dependent. Development of severe IgE-mediated hypovolemia and shock required VE-restricted ABL1 expression. Treatment of mice with a history of food-induced anaphylaxis with the ABL kinase inhibitor imatinib protected the mice from severe IgE-mediated anaphylaxis. CONCLUSION: IL-4 amplifies IgE- and histamine-induced VE dysfunction, fluid extravasation, and the severity of anaphylaxis through a VE IL-4Rα/ABL1-dependent mechanism. These studies implicate an important contribution by the VE compartment in the severity of anaphylaxis and identify a new pathway for therapeutic intervention of IgE-mediated reactions.


Assuntos
Anafilaxia/imunologia , Endotélio Vascular/imunologia , Imunoglobulina E/imunologia , Interleucina-4/administração & dosagem , Proteínas Proto-Oncogênicas c-abl/imunologia , Receptores de Interleucina-4/imunologia , Alérgenos/administração & dosagem , Alérgenos/imunologia , Animais , Anticorpos/administração & dosagem , Linhagem Celular , Feminino , Histamina/administração & dosagem , Humanos , Mesilato de Imatinib/farmacologia , Masculino , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Ovalbumina/administração & dosagem , Receptores de Interleucina-4/genética , Choque/imunologia
8.
Gastroenterology ; 153(1): 205-218.e8, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28390865

RESUMO

BACKGROUND & AIMS: The canonical Wnt signaling pathway activates the transcriptional activity of ß-catenin. This pathway is often activated in colorectal cancer cells, but strategies to block it in tumors have not been effective. The SAM pointed domain containing ETS transcription factor (SPDEF) suppresses formation of colon tumors by unclear mechanisms. We investigated these mechanisms and the effects of SPDEF on ß-catenin activity in mouse models of colorectal cancer (CRC), CRC cell lines, and mouse and human normal and cancer colonoids. METHODS: We performed studies of Lgr5CreERT2; ß-cateninexon3; Rosa26LSL-rtta-ires-EGFP; TRE-Spdef mice, which express an oncogenic form of ß-catenin in Lgr5-positive ISCs upon administration of tamoxifen and SPDEF upon administration of tetracycline. CRC lines (HCT116 and SW480) were engineered to express inducible tagged SPDEF or vector (control) and subcutaneously injected into immunodeficient NSG mice. We generated SPDEF-inducible human colonoids, including a line derived from normal rectal mucosa (control) and an adenocarcinoma line derived from a patient with germline MUTYH mutation. Full-length and truncated forms of SPDEF were expressed in CRC cells; cells were assayed for ß-catenin activity and studied in immunoprecipitation and chromatin immunoprecipitation assays. RESULTS: Expression of SPDEF was sufficient to inhibit intestinal tumorigenesis by activated ß-catenin, block tumor cell proliferation, and restrict growth of established tumors. In tumor cells with activated ß -catenin, expression of SPDEF induced a quiescent state, which was reversed when SPDEF expression was stopped. In mouse and human normal and tumor-derived enteroids/colonoids, those that expressed SPDEF for 3 days were significantly smaller. SPDEF inhibited the transcriptional activity of ß-catenin via a protein-protein interaction, independent of SPDEF DNA binding capacity. SPDEF disrupted ß-catenin binding to TCF1 and TCF3, displacing ß-catenin from enhancer regions of genes that regulate the cell cycle but not genes that regulate stem cell activities. CONCLUSIONS: In studies of mice and human CRC, we found that SPDEF induces a quiescent state in CRC cells by disrupting binding of ß-catenin to TCF1 and TCF3 and regulation of genes that control the cell cycle. In this model, ß-catenin activity determines the proliferation or quiescence of CRC cells based on the absence or presence of SPDEF.


Assuntos
Adenoma/genética , Neoplasias Colorretais/genética , Proteínas Proto-Oncogênicas c-ets/genética , Transcrição Gênica/genética , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo , Adenoma/química , Adenoma/metabolismo , Animais , Proteína Axina/genética , Carcinogênese , Ciclo Celular/genética , Proliferação de Células , Cromatina/metabolismo , Colo , Neoplasias Colorretais/química , Neoplasias Colorretais/metabolismo , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HEK293 , Humanos , Receptores de Hialuronatos/análise , Mucosa Intestinal/química , Mucosa Intestinal/patologia , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Masculino , Camundongos , Organoides , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-ets/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Receptor EphB2/genética , Células-Tronco , Fatores de Transcrição TCF/metabolismo , Transfecção
9.
Cell Mol Gastroenterol Hepatol ; 3(1): 51-71, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28174757

RESUMO

BACKGROUND & AIMS: The transcription factor atonal homolog 1 (ATOH1) controls the fate of intestinal progenitors downstream of the Notch signaling pathway. Intestinal progenitors that escape Notch activation express high levels of ATOH1 and commit to a secretory lineage fate, implicating ATOH1 as a gatekeeper for differentiation of intestinal epithelial cells. Although some transcription factors downstream of ATOH1, such as SPDEF, have been identified to specify differentiation and maturation of specific cell types, the bona fide transcriptional targets of ATOH1 still largely are unknown. Here, we aimed to identify ATOH1 targets and to identify transcription factors that are likely to co-regulate gene expression with ATOH1. METHODS: We used a combination of chromatin immunoprecipitation and messenger RNA-based high-throughput sequencing (ChIP-seq and RNA-seq), together with cell sorting and transgenic mice, to identify direct targets of ATOH1, and establish the epistatic relationship between ATOH1 and SPDEF. RESULTS: By using unbiased genome-wide approaches, we identified more than 700 genes as ATOH1 transcriptional targets in adult small intestine and colon. Ontology analysis indicated that ATOH1 directly regulates genes involved in specification and function of secretory cells. De novo motif analysis of ATOH1 targets identified SPDEF as a putative transcriptional co-regulator of ATOH1. Functional epistasis experiments in transgenic mice show that SPDEF amplifies ATOH1-dependent transcription but cannot independently initiate transcription of ATOH1 target genes. CONCLUSIONS: This study unveils the direct targets of ATOH1 in the adult intestines and illuminates the transcriptional events that initiate the specification and function of intestinal secretory lineages.

10.
Sci Rep ; 5: 17185, 2015 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-26597788

RESUMO

Leucine-rich repeat-containing G-protein coupled receptor 5-expressing (Lgr5(+)) cells have been identified as stem/progenitor cells in the circumvallate papillae, and single cultured Lgr5(+) cells give rise to taste cells. Here we use circumvallate papilla tissue to establish a three-dimensional culture system (taste bud organoids) that develops phenotypic characteristics similar to native tissue, including a multilayered epithelium containing stem/progenitor in the outer layers and taste cells in the inner layers. Furthermore, characterization of the cell cycle of the taste bud progenitor niche reveals striking dynamics of taste bud development and regeneration. Using this taste bud organoid culture system and FUCCI2 transgenic mice, we identify the stem/progenitor cells have at least 5 distinct cell cycle populations by tracking within 24-hour synchronized oscillations of proliferation. Additionally, we demonstrate that stem/progenitor cells have motility to form taste bud organoids. Taste bud organoids provides a system for elucidating mechanisms of taste signaling, disease modeling, and taste tissue regeneration.


Assuntos
Células-Tronco/fisiologia , Papilas Gustativas/citologia , Animais , Ciclo Celular , Proliferação de Células , Rastreamento de Células , Receptores de Hialuronatos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Organoides/citologia , Receptores Acoplados a Proteínas G/metabolismo , Técnicas de Cultura de Tecidos
11.
Stem Cell Reports ; 4(2): 209-25, 2015 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-25579133

RESUMO

Intestinal epithelial stem cells (IESCs) control the intestinal homeostatic response to inflammation and regeneration. The underlying mechanisms are unclear. Cytokine-STAT5 signaling regulates intestinal epithelial homeostasis and responses to injury. We link STAT5 signaling to IESC replenishment upon injury by depletion or activation of Stat5 transcription factor. We found that depletion of Stat5 led to deregulation of IESC marker expression and decreased LGR5(+) IESC proliferation. STAT5-deficient mice exhibited worse intestinal histology and impaired crypt regeneration after γ-irradiation. We generated a transgenic mouse model with inducible expression of constitutively active Stat5. In contrast to Stat5 depletion, activation of STAT5 increased IESC proliferation, accelerated crypt regeneration, and conferred resistance to intestinal injury. Furthermore, ectopic activation of STAT5 in mouse or human stem cells promoted LGR5(+) IESC self-renewal. Accordingly, STAT5 promotes IESC proliferation and regeneration to mitigate intestinal inflammation. STAT5 is a functional therapeutic target to improve the IESC regenerative response to gut injury.


Assuntos
Diferenciação Celular , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Regeneração , Fator de Transcrição STAT5/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Diferenciação Celular/genética , Proliferação de Células , Colite/etiologia , Colite/patologia , Modelos Animais de Doenças , Marcação de Genes , Loci Gênicos , Vetores Genéticos/genética , Humanos , Mucosa Intestinal/patologia , Mucosa Intestinal/efeitos da radiação , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Complexo Repressor Polycomb 1/genética , Ligação Proteica , Lesões por Radiação , Lesões Experimentais por Radiação , Tolerância a Radiação/genética , Regeneração/genética , Fator de Transcrição STAT5/genética , Ativação Transcricional
12.
Gastroenterology ; 144(5): 1012-1023.e6, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23376423

RESUMO

BACKGROUND & AIMS: Expression of the SAM pointed domain containing ETS transcription factor (SPDEF or prostate-derived ETS factor) is regulated by Atoh1 and is required for the differentiation of goblet and Paneth cells. SPDEF has been reported to suppress the development of breast, prostate, and colon tumors. We analyzed levels of SPDEF in colorectal tumor samples from patients and its tumor-suppressive functions in mouse models of colorectal cancer (CRC). METHODS: We analyzed levels of SPDEF messenger RNA and protein in more than 500 human CRC samples and more than 80 nontumor controls. Spdef(-/-)and wild-type mice (controls) were either bred with Apc(Min/+) mice, or given azoxymethane (AOM) and dextran sodium sulfate (DSS), or 1,2-dimethylhydrazine and DSS, to induce colorectal tumors. Expression of Spdef also was induced transiently by administration of tetracycline to Spdef(dox-intestine) mice with established tumors, induced by the combination of AOM and DSS or by breeding with Apc(Min/+) mice. Colon tissues were collected and analyzed for tumor number, size, grade, and for cell proliferation and apoptosis. We also analyzed the effects of SPDEF expression in HCT116 and SW480 human CRC cells. RESULTS: In colorectal tumors from patients, loss of SPDEF was observed in approximately 85% of tumors and correlated with progression from normal tissue, to adenoma, to adenocarcinoma. Spdef(-/-); Apc(Min/+) mice developed approximately 3-fold more colon tumors than Spdef(+/+); Apc(Min/+) mice. Likewise, Spdef(-/-) mice developed approximately 3-fold more colon tumors than Spdef(+/+) mice after administration of AOM and DSS. After administration of 1,2-dimethylhydrazine and DSS, invasive carcinomas were observed exclusively in Spdef(-/-) mice. Conversely, expression of SPDEF was sufficient to promote cell-cycle exit in cells of established adenomas from Spdef(dox-intestine); Apc(Min/+) mice and in Spdef(dox-intestine) mice after administration of AOM + DSS. SPDEF inhibited the expression of ß-catenin-target genes in mouse colon tumors, and interacted with ß-catenin to block its transcriptional activity in CRC cell lines, resulting in lower levels of cyclin D1 and c-MYC. CONCLUSIONS: SPDEF is a colon tumor suppressor and a candidate therapeutic target for colon adenomas and adenocarcinoma.


Assuntos
Adenocarcinoma/genética , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas c-ets/genética , RNA Neoplásico/genética , beta Catenina/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Apoptose , Proliferação de Células , Colo/metabolismo , Colo/patologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Progressão da Doença , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias Experimentais , Reação em Cadeia da Polimerase , Proteínas Proto-Oncogênicas c-ets/biossíntese , Análise Serial de Tecidos , Células Tumorais Cultivadas , beta Catenina/antagonistas & inibidores
13.
Annu Rev Physiol ; 75: 263-88, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23190077

RESUMO

The small and large intestines are tubular organs composed of several tissue types. The columnar epithelium that lines the inner surface of the intestines distinguishes the digestive physiology of each region of the intestine and consists of several distinct cell types that are rapidly and continually renewed by intestinal stem cells that reside near the base of the crypts of Lieberkühn. Notch signaling controls the fate of intestinal stem cells by regulating the expression of Hes genes and by repressing Atoh1. Alternate models of Notch pathway control of cell fate determination are presented. Roles for Notch signaling in development of the intestine, including mesenchymal and neural cells, are discussed. The oncogenic activities of Notch in colorectal cancer, as well as the tumor suppressive activities of Atoh1, are reviewed. Therapeutic targeting of the Notch pathway in colorectal cancers is discussed, along with potential caveats.


Assuntos
Homeostase/fisiologia , Intestinos/fisiologia , Receptores Notch/fisiologia , Animais , Transformação Celular Neoplásica , Neoplasias Colorretais/fisiopatologia , Humanos , Transdução de Sinais/fisiologia
14.
Exp Cell Res ; 317(19): 2702-10, 2011 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-21978911

RESUMO

In this review, we present an overview of intestinal development and cellular differentiation of the intestinal epithelium. The review is separated into two sections: Section one summarizes organogenesis of the small and large intestines, including endoderm and gut tube formation in early embryogenesis, villus morphogenesis, and crypt formation. Section two reviews cell fate specification and differentiation of each cell type within the intestinal epithelium. Growth factor and transcriptional networks that regulate these developmental processes are summarized.


Assuntos
Diferenciação Celular , Intestinos/embriologia , Intestinos/fisiologia , Animais , Padronização Corporal/fisiologia , Endoderma/embriologia , Endoderma/fisiologia , Humanos , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Intestinos/crescimento & desenvolvimento , Modelos Biológicos , Morfogênese/fisiologia
15.
J Biol Chem ; 286(15): 13357-69, 2011 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-21303908

RESUMO

Interleukin-13 (IL-13) has been linked to the pathogenesis of inflammatory diseases of the gastrointestinal tract. It is postulated that IL-13 drives inflammatory lesions through the modulation of both hematopoietic and nonhematopoietic cell function in the intestine. To delineate the relevant contribution of elevated levels of intestinal IL-13 to intestinal structure and function, we generated an intestinal IL-13 transgenic mouse (iIL-13Tg). We show that constitutive overexpression of IL-13 in the small bowel induces modification of intestinal epithelial architecture (villus blunting, goblet cell hyperplasia, and increased epithelial proliferation) and epithelial function (altered basolateral → apical Cl(-) ion conductance). Pharmacological analyses in vitro and in vivo determined that elevated Cl(-) conductance is mediated by altered cystic fibrosis transmembrane conductance regulator expression and activity. Generation of iIL-13Tg/Il13rα1(-/-), iIL-13Tg/Il13rα2(-/-), and iIL-13Tg/Stat6(-/-) mice revealed that IL-13-mediated dysregulation of epithelial architecture and Cl(-) conductance is dependent on IL-13Rα1 and STAT-6. These observations demonstrate a central role for the IL-13/IL-13Rα1 pathway in the regulation of intestinal epithelial cell Cl(-) secretion via up-regulation of cystic fibrosis transmembrane conductance regulator, suggesting an important role for this pathway in secretory diarrhea.


Assuntos
Cloretos/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Subunidade alfa1 de Receptor de Interleucina-13/metabolismo , Interleucina-13/metabolismo , Enteropatias/metabolismo , Mucosa Intestinal/metabolismo , Animais , Células CACO-2 , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Cistos/genética , Cistos/metabolismo , Cistos/patologia , Diarreia/genética , Diarreia/metabolismo , Diarreia/patologia , Fibrose , Humanos , Interleucina-13/genética , Subunidade alfa1 de Receptor de Interleucina-13/genética , Enteropatias/genética , Enteropatias/patologia , Mucosa Intestinal/patologia , Transporte de Íons/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Fator de Transcrição STAT6/genética , Fator de Transcrição STAT6/metabolismo
16.
Gastroenterology ; 140(4): 1219-1229.e1-2, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21262227

RESUMO

BACKGROUND & AIMS: GATA transcription factors regulate proliferation, differentiation, and gene expression in multiple organs. GATA4 is expressed in the proximal 85% of the small intestine and regulates the jejunal-ileal gradient in absorptive enterocyte gene expression. GATA6 is co-expressed with GATA4 but also is expressed in the ileum; its function in the mature small intestine is unknown. METHODS: We investigated the function of GATA6 in small intestine using adult mice with conditional, inducible deletion of Gata6, or Gata6 and Gata4, specifically in the intestine. RESULTS: In ileum, deletion of Gata6 caused a decrease in crypt cell proliferation and numbers of enteroendocrine and Paneth cells, an increase in numbers of goblet-like cells in crypts, and altered expression of genes specific to absorptive enterocytes. In contrast to ileum, deletion of Gata6 caused an increase in numbers of Paneth cells in jejunum and ileum. Deletion of Gata6 and Gata4 resulted in a jejunal and duodenal phenotype that was nearly identical to that in the ileum after deletion of Gata6 alone, revealing common functions for GATA6 and GATA4. CONCLUSIONS: GATA transcription factors are required for crypt cell proliferation, secretory cell differentiation, and absorptive enterocyte gene expression in the small intestinal epithelium.


Assuntos
Fator de Transcrição GATA4/genética , Fator de Transcrição GATA4/metabolismo , Fator de Transcrição GATA6/genética , Fator de Transcrição GATA6/metabolismo , Intestino Delgado/fisiologia , Animais , Contagem de Células , Diferenciação Celular/fisiologia , Divisão Celular/fisiologia , Duodeno/citologia , Duodeno/fisiologia , Enterócitos/citologia , Enterócitos/metabolismo , Enterócitos/fisiologia , Expressão Gênica/fisiologia , Íleo/citologia , Íleo/fisiologia , Absorção Intestinal/fisiologia , Mucosa Intestinal/citologia , Mucosa Intestinal/fisiologia , Intestino Delgado/citologia , Jejuno/citologia , Jejuno/fisiologia , Camundongos , Camundongos Transgênicos , Celulas de Paneth/citologia , Celulas de Paneth/metabolismo , Celulas de Paneth/fisiologia , Receptores Notch/metabolismo , Transdução de Sinais/fisiologia , Proteínas Wnt/metabolismo
17.
Gastroenterology ; 139(3): 918-28, 928.e1-6, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20621629

RESUMO

BACKGROUND & AIMS: The atonal homolog 1 (Atoh1) transcription factor is required for intestinal secretory (goblet, Paneth, enteroendocrine) cell differentiation. Notch/gamma-secretase inhibitors (GSIs) block proliferation and induce secretory cell differentiation in the intestine. We used genetic analyses of mice to determine whether Atoh1 mediates the effects of GSIs in normal and cancerous intestinal epithelia. METHODS: We studied mice with intestine-specific disruption of Atoh1 (Atoh1(Deltaintestine)), the adenomatosis polyposis coli (APC)(min) mutation, both mutations (Atoh1(Deltaintestine); APC(min)), or littermate controls; mice were given GSI or vehicle. Colorectal cancer (CRC) cell lines were treated with GSI or vehicle and with small hairpin RNAs to reduce ATOH1. Differentiation and homeostasis were assessed by protein, RNA, and histologic analyses. RESULTS: GSIs failed to induce secretory cell differentiation or apoptosis or decrease proliferation of Atoh1-null progenitor cells, compared with wild-type cells. Exposure of APC(min) adenomas to GSIs decreased proliferation and increased secretory cell numbers in an Atoh1-dependent manner. In CRC cells treated with GSI, ATOH1 levels were correlated inversely with proliferation. ATOH1 was required for secretory cell gene expression in cell lines and in mice. CONCLUSIONS: ATOH1 is required for all effects of GSIs in intestinal crypts and adenomas; Notch has no unique function in intestinal progenitors and cancer cells other than to regulate ATOH1 expression. Reducing ATOH1 activity might mitigate intestinal toxicity from systemic GSI therapy for nonintestinal diseases. Among gastrointestinal malignancies, ATOH1 mediates the effects of GSIs, so ATOH1 expression levels might predict responses to these inhibitors. We propose that only the subset of CRCs that retain ATOH1 expression will respond to GSIs.


Assuntos
Secretases da Proteína Precursora do Amiloide/antagonistas & inibidores , Antineoplásicos/farmacologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/metabolismo , Células Epiteliais/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Receptores Notch/antagonistas & inibidores , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Apoptose/efeitos dos fármacos , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Regulação Neoplásica da Expressão Gênica , Genes APC , Células HCT116 , Células HT29 , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Transgênicos , Interferência de RNA , RNA Mensageiro/metabolismo , Receptores Notch/metabolismo , Fatores de Tempo
18.
Exp Cell Res ; 316(3): 452-65, 2010 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19786015

RESUMO

BACKGROUND AND AIMS: SPDEF (also termed PDEF or PSE) is an ETS family transcription factor that regulates gene expression in the prostate and goblet cell hyperplasia in the lung. Spdef has been reported to be expressed in the intestine. In this paper, we identify an important role for Spdef in regulating intestinal epithelial cell homeostasis and differentiation. METHODS: SPDEF expression was inhibited in colon cancer cells to determine its ability to control goblet cell gene activation. The effects of transgenic expression of Spdef on intestinal differentiation and homeostasis were determined. RESULTS: In LS174T colon cancer cells treated with Notch/gamma-secretase inhibitor to activate goblet cell gene expression, shRNAs that inhibited SPDEF also repressed expression of goblet cell genes AGR2, MUC2, RETLNB, and SPINK4. Transgenic expression of Spdef caused the expansion of intestinal goblet cells and corresponding reduction in Paneth, enteroendocrine, and absorptive enterocytes. Spdef inhibited proliferation of intestinal crypt cells without induction of apoptosis. Prolonged expression of the Spdef transgene caused a progressive reduction in the number of crypts that expressed Spdef, consistent with its inhibitory effects on cell proliferation. CONCLUSIONS: Spdef was sufficient to inhibit proliferation of intestinal progenitors and induce differentiation into goblet cells; SPDEF was required for activation of goblet cell associated genes in vitro. These data support a model in which Spdef promotes terminal differentiation into goblet cells of a common goblet/Paneth progenitor.


Assuntos
Diferenciação Celular , Células Caliciformes/citologia , Células Caliciformes/metabolismo , Proteínas Proto-Oncogênicas c-ets/metabolismo , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/metabolismo , Doxiciclina/farmacologia , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células Caliciformes/efeitos dos fármacos , Humanos , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/ultraestrutura , Camundongos , Especificidade de Órgãos/efeitos dos fármacos , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-ets/genética , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Transgenes/genética
19.
J Virol ; 81(24): 13519-32, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17913803

RESUMO

Kaposi's sarcoma-associated herpesvirus/human herpesvirus 8 (HHV8) ORF50 encodes a transactivator, K-Rta, which functions as the switch from latent to lytic virus replication. K-bZIP interacts with K-Rta and can repress its transactivation activity for some viral promoters. Both K-Rta and K-bZIP are required for origin-dependent DNA replication. To determine the role of K-bZIP in the context of the viral genome, we generated a recombinant HHV8 bacterial artificial chromosome (BAC) with a deletion in the K-bZIP open reading frame. This BACmid, BAC36DeltaK8, displayed an enhanced growth phenotype with respect to virus production and accumulation of virus-encoded mRNAs measured by real-time PCR when K-Rta was used to induce the virus lytic cycle. Conversely, induction of the virus lytic cycle using tetradecanoyl phorbol acetate/n-butyrate resulted in no virus production and an aberrant gene expression pattern from BAC36DeltaK8-containing cells compared to wild-type (wt) BAC. This null virus phenotype was efficiently complemented by the expression of K-bZIP in trans, restoring virus production to wt BAC levels. Immunofluorescence staining revealed that subcellular localization of K-Rta was unchanged; however, a disruption of LANA subcellular localization was observed in cells harboring BAC36DeltaK8, suggesting that K-bZIP influences LANA localization. Coimmunoprecipitation experiments confirmed that K-bZIP interacts with LANA in BCBL-1 cells and in cotransfection assays. Lastly, the chromatin immunoprecipitation assay revealed that, in an environment where K-Rta is overexpressed and in the absence of K-bZIP, K-Rta binds to CAAT enhancer binding protein alpha sites within oriLyt, suggesting that it is K-Rta that supplies an essential replication function and that K-bZIP may serve to augment or facilitate the interaction of K-Rta with oriLyt.


Assuntos
Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Deleção de Genes , Herpesvirus Humano 8/fisiologia , Proteínas Imediatamente Precoces/metabolismo , Proteínas Repressoras/metabolismo , Transativadores/metabolismo , Regulação para Cima , Proteínas Virais/metabolismo , Replicação Viral , Antígenos Virais/metabolismo , Fatores de Transcrição de Zíper de Leucina Básica/genética , Linhagem Celular , Cromossomos Artificiais Bacterianos/genética , Regulação Viral da Expressão Gênica , Teste de Complementação Genética , Herpesvirus Humano 8/classificação , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/metabolismo , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Nucleares/metabolismo , Fenótipo , Recombinação Genética , Proteínas Repressoras/genética , Transativadores/genética , Transfecção , Proteínas Virais/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA