Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
J Transl Med ; 22(1): 337, 2024 Apr 08.
Artigo em Inglês | MEDLINE | ID: mdl-38589873

RESUMO

BACKGROUND: The mesenchymal subtype of colorectal cancer (CRC), associated with poor prognosis, is characterized by abundant expression of the cellular prion protein PrPC, which represents a candidate therapeutic target. How PrPC is induced in CRC remains elusive. This study aims to elucidate the signaling pathways governing PrPC expression and to shed light on the gene regulatory networks linked to PrPC. METHODS: We performed in silico analyses on diverse datasets of in vitro, ex vivo and in vivo models of mouse CRC and patient cohorts. We mined ChIPseq studies and performed promoter analysis. CRC cell lines were manipulated through genetic and pharmacological approaches. We created mice combining conditional inactivation of Apc in intestinal epithelial cells and overexpression of the human prion protein gene PRNP. Bio-informatic analyses were carried out in two randomized control trials totalizing over 3000 CRC patients. RESULTS: In silico analyses combined with cell-based assays identified the Wnt-ß-catenin and glucocorticoid pathways as upstream regulators of PRNP expression, with subtle differences between mouse and human. We uncover multiple feedback loops between PrPC and these two pathways, which translate into an aggravation of CRC pathogenesis in mouse. In stage III CRC patients, the signature defined by PRNP-CTNNB1-NR3C1, encoding PrPC, ß-catenin and the glucocorticoid receptor respectively, is overrepresented in the poor-prognosis, mesenchymal subtype and associates with reduced time to recurrence. CONCLUSIONS: An unleashed PrPC-dependent vicious circle is pathognomonic of poor prognosis, mesenchymal CRC. Patients from this aggressive subtype of CRC may benefit from therapies targeting the PRNP-CTNNB1-NR3C1 axis.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Humanos , Camundongos , Animais , Proteínas Priônicas/genética , Proteínas Priônicas/metabolismo , beta Catenina/metabolismo , Glucocorticoides , Neoplasias do Colo/genética , Neoplasias Colorretais/genética , Fenótipo , Prognóstico , Via de Sinalização Wnt , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral
2.
Cell Mol Gastroenterol Hepatol ; 15(3): 689-715, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36356835

RESUMO

BACKGROUND & AIMS: Axin1 is a negative regulator of wingless-type MMTV integration site family, member 1 (Wnt)/ß-catenin signaling with tumor-suppressor function. The Wnt pathway has a critical role in the intestine, both during homeostasis and cancer, but the role of Axin1 remains elusive. METHODS: We assessed the role of Axin1 in normal intestinal homeostasis, with control, epithelial-specific, Axin1-knockout mice (Axin1ΔIEC) and Axin2-knockout mice. We evaluated the tumor-suppressor function of Axin1 during chemically induced colorectal tumorigenesis and dextran sulfate sodium-induced colitis, and performed comparative gene expression profiling by whole-genome RNA sequencing. The clinical relevance of the Axin1-dependent gene expression signature then was tested in a database of 2239 clinical colorectal cancer (CRC) samples. RESULTS: We found that Axin1 was dispensable for normal intestinal homeostasis and redundant with Axin2 for Wnt pathway down-regulation. Axin1 deficiency in intestinal epithelial cells rendered mice more susceptible to chemically induced colon carcinogenesis, but reduced dextran sulfate sodium-induced colitis by attenuating the induction of a proinflammatory program. RNA-seq analyses identified an interferon γ/T-helper1 immune program controlled by Axin1 that enhances the inflammatory response and protects against CRC. The Axin1-dependent gene expression signature was applied to human CRC samples and identified a group of patients with potential vulnerability to immune checkpoint blockade therapies. CONCLUSIONS: Our study establishes, in vivo, that Axin1 has redundant function with Axin2 for Wnt down-regulation and infers a new role for Axin1. Physiologically, Axin1 stimulates gut inflammation via an interferon γ/Th1 program that prevents tumor growth. Linked to its T-cell-mediated effect, the colonic Axin1 signature offers therapeutic perspectives for CRC.


Assuntos
Colite , Interferon gama , Camundongos , Animais , Humanos , Sulfato de Dextrana/toxicidade , Carcinogênese/genética , Colite/induzido quimicamente , Via de Sinalização Wnt/genética , Camundongos Knockout , Proteína Axina/genética , Proteína Axina/metabolismo
3.
Biomaterials ; 282: 121380, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35101742

RESUMO

The field of intestinal biology is thirstily searching for different culture methods that complement the limitations of organoids, particularly the lack of a differentiated intestinal compartment. While being recognized as an important milestone for basic and translational biological studies, many primary cultures of intestinal epithelium (IE) rely on empirical trials using hydrogels of various stiffness, whose mechanical impact on epithelial organization remains vague until now. Here, we report the development of hydrogel scaffolds with a range of elasticities and their influence on IE expansion, organization, and differentiation. On stiff substrates (>5 kPa), mouse IE cells adopt a flat cell shape and detach in the short-term. In contrast, on soft substrates (80-500 Pa), they sustain for a long-term, pack into high density, develop columnar shape with improved apical-basal polarity and differentiation marker expression, a phenotype reminiscent of features in vivo mouse IE. We then developed a soft gel molding process to produce 3D Matrigel scaffolds of close-to-nature stiffness, which support and maintain a culture of mouse IE into crypt-villus architecture. Thus, the present work is up-to-date informative for the design of biomaterials for ex vivo intestinal models, offering self-renewal in vitro culture that emulates the mouse IE.


Assuntos
Biomimética , Intestinos , Animais , Diferenciação Celular , Hidrogéis/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Organoides
4.
Hepatol Commun ; 5(9): 1490-1506, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34510835

RESUMO

CTNNB1 (catenin beta 1)-mutated hepatocellular carcinomas (HCCs) account for a large proportion of human HCCs. They display high levels of respiratory chain activity. As metabolism and redox balance are closely linked, tumor cells must maintain their redox status during these metabolic alterations. We investigated the redox balance of these HCCs and the feasibility of targeting this balance as an avenue for targeted therapy. We assessed the expression of the nuclear erythroid 2 p45-related factor 2 (NRF2) detoxification pathway in an annotated human HCC data set and reported an enrichment of the NRF2 program in human HCCs with CTNNB1 mutations, largely independent of NFE2L2 (nuclear factor, erythroid 2 like 2) or KEAP1 (Kelch-like ECH-associated protein 1) mutations. We then used mice with hepatocyte-specific oncogenic ß-catenin activation to evaluate the redox status associated with ß-catenin activation in preneoplastic livers and tumors. We challenged them with various oxidative stressors and observed that the ß-catenin pathway activation increased transcription of Nfe2l2, which protects ß-catenin-activated hepatocytes from oxidative damage and supports tumor development. Moreover, outside of its effects on reactive oxygen species scavenging, we found out that Nrf2 itself contributes to the metabolic activity of ß-catenin-activated cells. We then challenged ß-catenin activated tumors pharmacologically to create a redox imbalance and found that pharmacological inactivation of Nrf2 was sufficient to considerably decrease the progression of ß-catenin-dependent HCC development. Conclusion: These results demonstrate cooperation between oncogenic ß-catenin signaling and the NRF2 pathway in CTNNB1-mediated HCC tumorigenesis, and we provide evidence for the relevance of redox balance targeting as a therapeutic strategy in CTNNB1-mutated HCC.

5.
Nature ; 594(7863): 430-435, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34079124

RESUMO

The tumour suppressor APC is the most commonly mutated gene in colorectal cancer. Loss of Apc in intestinal stem cells drives the formation of adenomas in mice via increased WNT signalling1, but reduced secretion of WNT ligands increases the ability of Apc-mutant intestinal stem cells to colonize a crypt (known as fixation)2. Here we investigated how Apc-mutant cells gain a clonal advantage over wild-type counterparts to achieve fixation. We found that Apc-mutant cells are enriched for transcripts that encode several secreted WNT antagonists, with Notum being the most highly expressed. Conditioned medium from Apc-mutant cells suppressed the growth of wild-type organoids in a NOTUM-dependent manner. Furthermore, NOTUM-secreting Apc-mutant clones actively inhibited the proliferation of surrounding wild-type crypt cells and drove their differentiation, thereby outcompeting crypt cells from the niche. Genetic or pharmacological inhibition of NOTUM abrogated the ability of Apc-mutant cells to expand and form intestinal adenomas. We identify NOTUM as a key mediator during the early stages of mutation fixation that can be targeted to restore wild-type cell competitiveness and provide preventative strategies for people at a high risk of developing colorectal cancer.


Assuntos
Competição entre as Células , Transformação Celular Neoplásica , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Esterases/metabolismo , Genes APC , Mutação , Adenoma/genética , Adenoma/patologia , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Competição entre as Células/genética , Diferenciação Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Meios de Cultivo Condicionados , Progressão da Doença , Esterases/antagonistas & inibidores , Esterases/genética , Feminino , Humanos , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Organoides/citologia , Organoides/metabolismo , Organoides/patologia , Células-Tronco/citologia , Células-Tronco/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt
7.
Nat Commun ; 11(1): 6127, 2020 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-33257663

RESUMO

Excessive glucose production by the liver is a key factor in the hyperglycemia observed in type 2 diabetes mellitus (T2DM). Here, we highlight a novel role of liver kinase B1 (Lkb1) in this regulation. We show that mice with a hepatocyte-specific deletion of Lkb1 have higher levels of hepatic amino acid catabolism, driving gluconeogenesis. This effect is observed during both fasting and the postprandial period, identifying Lkb1 as a critical suppressor of postprandial hepatic gluconeogenesis. Hepatic Lkb1 deletion is associated with major changes in whole-body metabolism, leading to a lower lean body mass and, in the longer term, sarcopenia and cachexia, as a consequence of the diversion of amino acids to liver metabolism at the expense of muscle. Using genetic, proteomic and pharmacological approaches, we identify the aminotransferases and specifically Agxt as effectors of the suppressor function of Lkb1 in amino acid-driven gluconeogenesis.


Assuntos
Aminoácidos/metabolismo , Gluconeogênese/fisiologia , Fígado/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP , Animais , Caquexia , Diabetes Mellitus Tipo 2/metabolismo , Jejum , Feminino , Glucose/metabolismo , Hepatócitos/metabolismo , Hiperglicemia/metabolismo , Masculino , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/genética , Proteômica , Sarcopenia , Transaminases/metabolismo
8.
Cell Death Differ ; 27(10): 2872-2887, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32355182

RESUMO

Ribosome biogenesis inhibition causes cell cycle arrest and apoptosis through the activation of tumor suppressor-dependent surveillance pathways. These responses are exacerbated in cancer cells, suggesting that targeting ribosome synthesis may be beneficial to patients. Here, we characterize the effect of the loss-of-function of Notchless (Nle), an essential actor of ribosome biogenesis, on the intestinal epithelium undergoing tumor initiation due to acute Apc loss-of-function. We show that ribosome biogenesis dysfunction strongly alleviates Wnt-driven tumor initiation by restoring cell cycle exit and differentiation in Apc-deficient progenitors. Conversely Wnt hyperactivation attenuates the cellular responses to surveillance pathways activation induced by ribosome biogenesis dysfunction, as proliferation was maintained at control-like levels in the stem cells and progenitors of double mutants. Thus, our data indicate that, while ribosome biogenesis inhibition efficiently reduces cancer cell proliferation in the intestinal epithelium, enhanced resistance of Apc-deficient stem and progenitor cells to ribosome biogenesis defects may be an important concern when using a therapeutic strategy targeting ribosome production for the treatment of Wnt-dependent tumorigenesis.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Transformação Celular Neoplásica , Mucosa Intestinal , Proteínas de Membrana/fisiologia , Ribossomos/metabolismo , Via de Sinalização Wnt , Animais , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Biogênese de Organelas
9.
Proc Natl Acad Sci U S A ; 117(20): 11136-11146, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32371487

RESUMO

The intestinal epithelium acts as a barrier between the organism and its microenvironment, including the gut microbiota. It is the most rapidly regenerating tissue in the human body thanks to a pool of intestinal stem cells (ISCs) expressing Lgr5 The intestinal epithelium has to cope with continuous stress linked to its digestive and barrier functions. Epithelial repair is crucial to maintain its integrity, and Lgr5-positive intestinal stem cell (Lgr5+ISC) resilience following cytotoxic stresses is central to this repair stage. We show here that autophagy, a pathway allowing the lysosomal degradation of intracellular components, plays a crucial role in the maintenance and genetic integrity of Lgr5+ISC under physiological and stress conditions. Using conditional mice models lacking the autophagy gene Atg7 specifically in all intestinal epithelial cells or in Lgr5+ISC, we show that loss of Atg7 induces the p53-mediated apoptosis of Lgr5+ISC. Mechanistically, this is due to increasing oxidative stress, alterations to interactions with the microbiota, and defective DNA repair. Following irradiation, we show that Lgr5+ISC repair DNA damage more efficiently than their progenitors and that this protection is Atg7 dependent. Accordingly, we found that the stimulation of autophagy on fasting protects Lgr5+ISC against DNA damage and cell death mediated by oxaliplatin and doxorubicin treatments. Finally, p53 deletion prevents the death of Atg7-deficient Lgr5+ISC but promotes genetic instability and tumor formation. Altogether, our findings provide insights into the mechanisms underlying maintenance and integrity of ISC and highlight the key functions of Atg7 and p53.


Assuntos
Proteína 7 Relacionada à Autofagia/metabolismo , Autofagia/fisiologia , Intestinos/fisiologia , Células-Tronco/metabolismo , Animais , Apoptose , Proteína 7 Relacionada à Autofagia/genética , Dano ao DNA , Reparo do DNA , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Feminino , Genes p53/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Intestinos/patologia , Masculino , Camundongos , Camundongos Knockout , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/citologia
10.
J Hepatol ; 73(3): 628-639, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32205193

RESUMO

BACKGROUND & AIMS: Paneth cells (PCs) synthesize and secrete antimicrobial peptides that are key mediators of host-microbe interactions, establishing a balance between intestinal microflora and enteric pathogens. We observed that their number increases in experimental portal hypertension and aimed to investigate the mechanisms by which these cells can contribute to the regulation of portal pressure. METHODS: We first treated Math1Lox/LoxVilcreERT2 mice with tamoxifen to induce the complete depletion of intestinal PCs. Subsequently, we performed partial portal vein or bile duct ligation. We then studied the effects of these interventions on hemodynamic parameters, proliferation of blood vessels and the expression of genes regulating angiogenesis. Intestinal organoids were cultured and exposed to different microbial products to study the composition of their secreted products (by proteomics) and their effects on the proliferation and tube formation of endothelial cells (ECs). In vivo confocal laser endomicroscopy was used to confirm the findings on blood vessel proliferation. RESULTS: Portal hypertension was significantly attenuated in PC-depleted mice compared to control mice and was associated with a decrease in portosystemic shunts. Depletion of PCs also resulted in a significantly decreased density of blood vessels in the intestinal wall and mesentery. Furthermore, we observed reduced expression of intestinal genes regulating angiogenesis in Paneth cell depleted mice using arrays and next generation sequencing. Tube formation and wound healing responses were significantly decreased in ECs treated with conditioned media from PC-depleted intestinal organoids exposed to intestinal microbiota-derived products. Proteomic analysis of conditioned media in the presence of PCs revealed an increase in factors regulating angiogenesis and additional metabolic processes. In vivo endomicroscopy showed decreased vascular proliferation in the absence of PCs. CONCLUSIONS: These results suggest that in response to intestinal flora and microbiota-derived factors, PCs secrete not only antimicrobial peptides, but also pro-angiogenic signaling molecules, thereby promoting intestinal and mesenteric angiogenesis and regulating portal hypertension. LAY SUMMARY: Paneth cells are present in the lining of the small intestine. They prevent the passage of bacteria from the intestine into the blood circulation by secreting substances to fight bacteria. In this paper, we discovered that these substances not only act against bacteria, but also increase the quantity of blood vessels in the intestine and blood pressure in the portal vein. This is important, because high blood pressure in the portal vein may result in several complications which could be targeted with novel approaches.


Assuntos
Infecções por Escherichia coli/metabolismo , Escherichia coli/metabolismo , Microbioma Gastrointestinal/genética , Hipertensão Portal/metabolismo , Hipertensão Portal/microbiologia , Neovascularização Patológica/metabolismo , Celulas de Paneth/metabolismo , Animais , Meios de Cultivo Condicionados , Modelos Animais de Doenças , Infecções por Escherichia coli/microbiologia , Células Endoteliais da Veia Umbilical Humana/metabolismo , Humanos , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Masculino , Camundongos , Camundongos Transgênicos , Organoides/metabolismo , Organoides/microbiologia , Celulas de Paneth/efeitos dos fármacos , Proteínas Citotóxicas Formadoras de Poros/metabolismo , Proteoma , Proteômica/métodos , Tamoxifeno/farmacologia
11.
J Pathol ; 250(3): 251-261, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31729028

RESUMO

Traditional serrated adenoma (TSA) remains the least understood of all the colorectal adenomas, although these lesions have been associated with a significant cancer risk, twice that of the conventional adenoma (CAD) and of the sessile serrated adenoma (SSA/P). This study was performed to investigate the proteomic profiles of the different colorectal adenomas to better understand the pathogenesis of TSA. We performed a global quantitative proteome analysis using the label-free quantification (LFQ) method on 44 colorectal adenoma (12 TSAs, 15 CADs, and 17 SSA/Ps) and 17 normal colonic mucosa samples, archived as formalin-fixed paraffin-embedded blocks. Unsupervised consensus hierarchical clustering applied to the whole proteomic profile of the 44 colorectal adenomas identified four subtypes: C1 and C2 were well-individualized clusters composed of all the CADs (15/15) and most of the SSA/Ps (13/17), respectively. This is consistent with the fact that CADs and SSA/Ps are homogeneous and distinct colorectal adenoma entities. In contrast, TSAs were subdivided into C3 and C4 clusters, consistent with the more heterogeneous entity of TSA at the morphologic and molecular levels. Comparison of the proteome expression profile between the adenoma subtypes and normal colonic mucosa further confirmed the heterogeneous nature of TSAs, which overlapped either on CADs or SSA/Ps, whereas CADs and SSAs formed homogeneous and distinct entities. Furthermore, we identified LEFTY1 a new potential marker for TSAs that may be relevant for the pathogenesis of TSA. LEFTY1 is an inhibitor of the Nodal/TGFß pathway, which we found to be one of the most overexpressed proteins specifically in TSAs. This finding was confirmed by immunohistochemistry. Our study confirms that CADs and SSA/Ps form homogeneous and distinct colorectal adenoma entities, whereas TSAs are a heterogeneous entity and may arise from either SSA/Ps or from normal mucosa evolving through a process related to the CAD pathway. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Assuntos
Adenoma/metabolismo , Colo/metabolismo , Neoplasias Colorretais/metabolismo , Proteoma , Adenoma/patologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Colo/patologia , Neoplasias Colorretais/patologia , Feminino , Humanos , Imuno-Histoquímica , Masculino , Espectrometria de Massas , Pessoa de Meia-Idade , Inclusão em Parafina , Proteômica
12.
Med Sci (Paris) ; 33(3): 290-296, 2017 Mar.
Artigo em Francês | MEDLINE | ID: mdl-28367816

RESUMO

The intestinal epithelial cells are crucial mediators of intestinal homeostasis. The intestinal epithelium is the largest of the body's mucosal surfaces exposed to the environment. Intestinal homeostasis is essentially based on the maintenance of intestinal epithelial cell integrity, a complex process involving a balance between the intestinal flora, the immune system and the energy expenses linked to metabolism. Autophagy appears to be central to these functions and allows the epithelium to adapt to its environment and different stress situations by participating in antibacterial defense, by controlling the composition of the intestinal flora and the immune response, and by participating in energy homeostasis. Alterations of this protective mechanism are involved in inflammatory bowel diseases and colorectal cancer.


Assuntos
Autofagia/fisiologia , Homeostase/fisiologia , Intestinos/fisiologia , Animais , Neoplasias Colorretais/patologia , Neoplasias Colorretais/fisiopatologia , Humanos , Doenças Inflamatórias Intestinais/patologia , Doenças Inflamatórias Intestinais/fisiopatologia , Enteropatias/patologia , Enteropatias/fisiopatologia , Mucosa Intestinal/patologia , Mucosa Intestinal/fisiologia , Intestinos/patologia
15.
Nat Cell Biol ; 17(8): 1062-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26214133

RESUMO

Here, we show that autophagy is activated in the intestinal epithelium in murine and human colorectal cancer and that the conditional inactivation of Atg7 in intestinal epithelial cells inhibits the formation of pre-cancerous lesions in Apc(+/-) mice by enhancing anti-tumour responses. The antibody-mediated depletion of CD8(+) T cells showed that these cells are essential for the anti-tumoral responses mediated by the inhibition of autophagy. We show that Atg7 deficiency leads to intestinal dysbiosis and that the microbiota is required for anticancer responses. In addition, Atg7 deficiency resulted in a stress response accompanied by metabolic defects, AMPK activation and p53-mediated cell-cycle arrest in tumour cells but not in normal tissue. This study reveals that the inhibition of autophagy within the epithelium may prevent the development and progression of colorectal cancer in genetically predisposed patients.


Assuntos
Adenocarcinoma/prevenção & controle , Adenoma/prevenção & controle , Transformação Celular Neoplásica/metabolismo , Colo/metabolismo , Neoplasias Colorretais/prevenção & controle , Imunidade nas Mucosas , Microbiota/imunologia , Proteínas Associadas aos Microtúbulos/deficiência , Enzimas Ativadoras de Ubiquitina/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Adenocarcinoma/microbiologia , Adenocarcinoma/patologia , Adenoma/genética , Adenoma/imunologia , Adenoma/microbiologia , Adenoma/patologia , Animais , Autofagia , Proteína 7 Relacionada à Autofagia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/microbiologia , Pontos de Checagem do Ciclo Celular , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/imunologia , Transformação Celular Neoplásica/patologia , Colo/imunologia , Colo/microbiologia , Colo/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/patologia , Modelos Animais de Doenças , Disbiose , Ativação Enzimática , Feminino , Genes APC , Interações Hospedeiro-Patógeno , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Associadas aos Microtúbulos/genética , Fatores de Tempo , Carga Tumoral , Proteína Supressora de Tumor p53/metabolismo
16.
J Cell Biol ; 198(3): 331-41, 2012 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-22851318

RESUMO

The stem cells (SCs) at the bottom of intestinal crypts tightly contact niche-supporting cells and fuel the extraordinary tissue renewal of intestinal epithelia. Their fate is regulated stochastically by populational asymmetry, yet whether asymmetrical fate as a mode of SC division is relevant and whether the SC niche contains committed progenitors of the specialized cell types are under debate. We demonstrate spindle alignments and planar cell polarities, which form a novel functional unit that, in SCs, can yield daughter cell anisotropic movement away from niche-supporting cells. We propose that this contributes to SC homeostasis. Importantly, we demonstrate that some SC divisions are asymmetric with respect to cell fate and provide data suggesting that, in some SCs, mNumb displays asymmetric segregation. Some of these processes were altered in apparently normal crypts and microadenomas of mice carrying germline Apc mutations, shedding new light on the first stages of progression toward colorectal cancer.


Assuntos
Proteína da Polipose Adenomatosa do Colo/fisiologia , Mucosa Intestinal/metabolismo , Actinas/química , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Anisotropia , Linhagem Celular , Cromatina/química , Cruzamentos Genéticos , Progressão da Doença , Cães , Homeostase , Interfase , Intestinos/patologia , Camundongos , Camundongos Knockout , Microscopia Confocal/métodos , Mutação , Processos Estocásticos , Telófase
17.
Proc Natl Acad Sci U S A ; 109(23): 8965-70, 2012 Jun 05.
Artigo em Inglês | MEDLINE | ID: mdl-22586121

RESUMO

Intestinal epithelium has the capacity to self-renew and generate differentiated cells through the existence of two types of epithelial stem cells: active crypt base columnar cells (CBCs) and quiescent +4 cells. The behaviors of these cells are regulated both by intrinsic programs and by extrinsic signals sent by neighboring cells, which define the niche. It is clear that the ß-catenin pathway acts as an essential intrinsic signal for the maintenance and proliferation of CBC, and it was recently proposed that Paneth cells provide a crucial niche by secreting Wingless/Int (Wnt) ligands. Here, we examined the effect of disrupting the intestinal stem cell niche by inducible deletion of the transcription factor Math1 (Atoh1), an essential driver of secretory cell differentiation. We found that complete loss of Paneth cells attributable to Math1 deficiency did not perturb the crypt architecture and allowed the maintenance and proliferation of CBCs. Indeed, Math1-deficient crypt cells tolerated in vivo Paneth cell loss and maintained active ß-catenin signaling but could not grow ex vivo without exogenous Wnt, implying that, in vivo, underlying mucosal cells act as potential niche. Upon irradiation, Math1-deficient crypt cells regenerated and CBCs continued cycling. Finally, CBC stem cells deficient in adenomatous polyposis coli (Apc) and Math1 were able to promote intestinal tumorigenesis. We conclude that in vivo, Math1-deficient crypts counteract the absence of Paneth cell-derived Wnts and prevent CBC stem cell exhaustion.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Mucosa Intestinal/citologia , Celulas de Paneth/citologia , Transdução de Sinais/fisiologia , Células-Tronco/ultraestrutura , beta Catenina/metabolismo , Animais , Imuno-Histoquímica , Hibridização In Situ , Camundongos , Análise em Microsséries , Microscopia Eletrônica , Reação em Cadeia da Polimerase , Proteínas Wnt/deficiência
19.
J Cell Biol ; 192(5): 767-80, 2011 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-21383077

RESUMO

The unique morphology of tuft cells was first revealed by electron microscopy analyses in several endoderm-derived epithelia. Here, we explore the relationship of these cells with the other cell types of the intestinal epithelium and describe the first marker signature allowing their unambiguous identification. We demonstrate that although mature tuft cells express DCLK1, a putative marker of quiescent stem cells, they are post-mitotic, short lived, derive from Lgr5-expressing epithelial stem cells, and are found in mouse and human tumors. We show that whereas the ATOH1/MATH1 transcription factor is essential for their differentiation, Neurog3, SOX9, GFI1, and SPDEF are dispensable, which distinguishes these cells from enteroendocrine, Paneth, and goblet cells, and raises from three to four the number of secretory cell types in the intestinal epithelium. Moreover, we show that tuft cells are the main source of endogenous intestinal opioids and are the only epithelial cells that express cyclooxygenase enzymes, suggesting important roles for these cells in the intestinal epithelium physiopathology.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mucosa Intestinal/citologia , Proteínas do Tecido Nervoso/metabolismo , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenoma/metabolismo , Adenoma/patologia , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/fisiologia , Biomarcadores/análise , Biomarcadores/metabolismo , Diferenciação Celular , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Quinases Semelhantes a Duplacortina , Humanos , Mucosa Intestinal/metabolismo , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Intestino Grosso/citologia , Intestino Grosso/metabolismo , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Oxirredutases Intramoleculares , Isomerases/metabolismo , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/fisiologia , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição SOX9/metabolismo
20.
Gut ; 60(2): 166-76, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21205878

RESUMO

AIMS: The activation of ß-catenin signalling is a key step in intestinal tumorigenesis. Interplay between the ß-catenin and Notch pathways during tumorigenesis has been reported, but the mechanisms involved and the role of Notch remain unclear. METHODS: Notch status was analysed by studying expression of the Notch effector Hes1 and Notch ligands/receptors in human colorectal cancer (CRC) and mouse models of Apc mutation. A genetic approach was used, deleting the Apc and RBP-J or Atoh1 genes in murine intestine. CRC cell lines were used to analyse the control of Hes1 and Atoh1 by ß-catenin signalling. RESULTS: Notch signalling was found to be activated downstream from ß-catenin. It was rapidly induced and maintained throughout tumorigenesis. Hes1 induction was mediated by ß-catenin and resulted from both the induction of the Notch ligand/receptor and Notch-independent control of the Hes1 promoter by ß-catenin. Surprisingly, the strong phenotype of unrestricted proliferation and impaired differentiation induced by acute Apc deletion in the intestine was not rescued by conditional Notch inactivation. Hyperactivation of ß-catenin signalling overrode the forced differention induced by Notch inhibition, through the downregulation of Atoh1, a key secretory determinant factor downstream of Notch. This process involves glycogen synthase kinase 3 ß (GSK3ß) and proteasome-mediated degradation. The restoration of Atoh1 expression in CRC cell lines displaying ß-catenin activation was sufficient to increase goblet cell differentiation, whereas genetic ablation of Atoh1 greatly increased tumour formation in Apc mutant mice. CONCLUSION: Notch signalling is a downstream target of ß-catenin hyperactivation in intestinal tumorigenesis. However, its inhibition had no tumour suppressor effect in the context of acute ß-catenin activation probably due to the downregulation of Atoh1. This finding calls into question the use of γ-secretase inhibitors for the treatment of CRC and suggests that the restoration of Atoh1 expression in CRC should be considered as a therapeutic approach.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Transformação Celular Neoplásica/metabolismo , Neoplasias Colorretais/metabolismo , Proteínas de Homeodomínio/metabolismo , Receptores Notch/metabolismo , beta Catenina/metabolismo , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/deficiência , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Análise Mutacional de DNA/métodos , DNA de Neoplasias/genética , Genes APC , Genes Neoplásicos , Humanos , Proteína de Ligação a Sequências Sinal de Recombinação J de Imunoglobina/deficiência , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Interferência de RNA , Receptores Notch/genética , Transdução de Sinais/fisiologia , Fatores de Transcrição HES-1
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA