Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Cell ; 152(1-2): 25-38, 2013 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-23273993

RESUMO

Cell-type plasticity within a tumor has recently been suggested to cause a bidirectional conversion between tumor-initiating stem cells and nonstem cells triggered by an inflammatory stroma. NF-κB represents a key transcription factor within the inflammatory tumor microenvironment. However, NF-κB's function in tumor-initiating cells has not been examined yet. Using a genetic model of intestinal epithelial cell (IEC)-restricted constitutive Wnt-activation, which comprises the most common event in the initiation of colon cancer, we demonstrate that NF-κB modulates Wnt signaling and show that IEC-specific ablation of RelA/p65 retards crypt stem cell expansion. In contrast, elevated NF-κB signaling enhances Wnt activation and induces dedifferentiation of nonstem cells that acquire tumor-initiating capacity. Thus, our data support the concept of bidirectional conversion and highlight the importance of inflammatory signaling for dedifferentiation and generation of tumor-initiating cells in vivo.


Assuntos
Desdiferenciação Celular , Transformação Celular Neoplásica , Neoplasias do Colo/patologia , Células-Tronco Neoplásicas/patologia , Animais , Colo/patologia , Células Epiteliais/patologia , Feminino , Humanos , Masculino , Camundongos , NF-kappa B/metabolismo , Via de Sinalização Wnt
2.
Nature ; 607(7919): 548-554, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35831497

RESUMO

The morphology and functionality of the epithelial lining differ along the intestinal tract, but tissue renewal at all sites is driven by stem cells at the base of crypts1-3. Whether stem cell numbers and behaviour vary at different sites is unknown. Here we show using intravital microscopy that, despite similarities in the number and distribution of proliferative cells with an Lgr5 signature in mice, small intestinal crypts contain twice as many effective stem cells as large intestinal crypts. We find that, although passively displaced by a conveyor-belt-like upward movement, small intestinal cells positioned away from the crypt base can function as long-term effective stem cells owing to Wnt-dependent retrograde cellular movement. By contrast, the near absence of retrograde movement in the large intestine restricts cell repositioning, leading to a reduction in effective stem cell number. Moreover, after suppression of the retrograde movement in the small intestine, the number of effective stem cells is reduced, and the rate of monoclonal conversion of crypts is accelerated. Together, these results show that the number of effective stem cells is determined by active retrograde movement, revealing a new channel of stem cell regulation that can be experimentally and pharmacologically manipulated.


Assuntos
Contagem de Células , Movimento Celular , Intestinos , Células-Tronco , Animais , Mucosa Intestinal/citologia , Intestino Delgado/citologia , Intestinos/citologia , Camundongos , Receptores Acoplados a Proteínas G , Células-Tronco/citologia , Proteínas Wnt
3.
Nature ; 517(7535): 497-500, 2015 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-25383520

RESUMO

Inactivation of APC is a strongly predisposing event in the development of colorectal cancer, prompting the search for vulnerabilities specific to cells that have lost APC function. Signalling through the mTOR pathway is known to be required for epithelial cell proliferation and tumour growth, and the current paradigm suggests that a critical function of mTOR activity is to upregulate translational initiation through phosphorylation of 4EBP1 (refs 6, 7). This model predicts that the mTOR inhibitor rapamycin, which does not efficiently inhibit 4EBP1 (ref. 8), would be ineffective in limiting cancer progression in APC-deficient lesions. Here we show in mice that mTOR complex 1 (mTORC1) activity is absolutely required for the proliferation of Apc-deficient (but not wild-type) enterocytes, revealing an unexpected opportunity for therapeutic intervention. Although APC-deficient cells show the expected increases in protein synthesis, our study reveals that it is translation elongation, and not initiation, which is the rate-limiting component. Mechanistically, mTORC1-mediated inhibition of eEF2 kinase is required for the proliferation of APC-deficient cells. Importantly, treatment of established APC-deficient adenomas with rapamycin (which can target eEF2 through the mTORC1-S6K-eEF2K axis) causes tumour cells to undergo growth arrest and differentiation. Taken together, our data suggest that inhibition of translation elongation using existing, clinically approved drugs, such as the rapalogs, would provide clear therapeutic benefit for patients at high risk of developing colorectal cancer.


Assuntos
Transformação Celular Neoplásica/patologia , Neoplasias Intestinais/metabolismo , Neoplasias Intestinais/patologia , Complexos Multiproteicos/metabolismo , Elongação Traducional da Cadeia Peptídica , Serina-Treonina Quinases TOR/metabolismo , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Quinase do Fator 2 de Elongação/deficiência , Quinase do Fator 2 de Elongação/genética , Quinase do Fator 2 de Elongação/metabolismo , Ativação Enzimática , Genes APC , Neoplasias Intestinais/genética , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica p55(v-myc)/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais , Proteínas Wnt/metabolismo
4.
EMBO J ; 34(18): 2321-33, 2015 Sep 14.
Artigo em Inglês | MEDLINE | ID: mdl-26240067

RESUMO

Wnt pathway deregulation is a common characteristic of many cancers. Only colorectal cancer predominantly harbours mutations in APC, whereas other cancer types (hepatocellular carcinoma, solid pseudopapillary tumours of the pancreas) have activating mutations in ß-catenin (CTNNB1). We have compared the dynamics and the potency of ß-catenin mutations in vivo. Within the murine small intestine (SI), an activating mutation of ß-catenin took much longer to achieve Wnt deregulation and acquire a crypt-progenitor cell (CPC) phenotype than Apc or Gsk3 loss. Within the colon, a single activating mutation of ß-catenin was unable to drive Wnt deregulation or induce the CPC phenotype. This ability of ß-catenin mutation to differentially transform the SI versus the colon correlated with higher expression of E-cadherin and a higher number of E-cadherin:ß-catenin complexes at the membrane. Reduction in E-cadherin synergised with an activating mutation of ß-catenin resulting in a rapid CPC phenotype within the SI and colon. Thus, there is a threshold of ß-catenin that is required to drive transformation, and E-cadherin can act as a buffer to sequester mutated ß-catenin.


Assuntos
Caderinas/metabolismo , Transformação Celular Neoplásica , Neoplasias do Colo , Mutação , Proteínas de Neoplasias , Via de Sinalização Wnt , beta Catenina , Animais , Caderinas/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Neoplasias do Colo/genética , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
5.
STAR Protoc ; 3(1): 101050, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-34977689

RESUMO

Intestinal organoid cultures are a powerful tool to study epithelial cells in vitro, as they are able to proliferate and differentiate into all cell lineages observed in vivo. Co-culturing organoids with distinct genetic backgrounds provides an excellent approach to study contact dependent and independent interactions between healthy and mutant epithelial intestinal cells. Here, we provide 2D and 3D approaches to mouse organoid co-cultures using fluorescently labeled organoids and demonstrate the analysis of these co-cultures using flow cytometry and microscopy-based approaches. For complete details on the use and execution of this profile, please refer to van Neerven et al., 2021.


Assuntos
Competição entre as Células , Organoides , Animais , Técnicas de Cocultura , Células Epiteliais , Intestinos , Camundongos
6.
Biol Open ; 11(12)2022 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-36350252

RESUMO

The rapid renewal of the epithelial gut lining is fuelled by stem cells that reside at the base of intestinal crypts. The signal transduction pathways and morphogens that regulate intestinal stem cell self-renewal and differentiation have been extensively characterised. In contrast, although extracellular matrix (ECM) components form an integral part of the intestinal stem cell niche, their direct influence on the cellular composition is less well understood. We set out to systematically compare the effect of two ECM classes, the interstitial matrix and the basement membrane, on the intestinal epithelium. We found that both collagen I and laminin-containing cultures allow growth of small intestinal epithelial cells with all cell types present in both cultures, albeit at different ratios. The collagen cultures contained a subset of cells enriched in fetal-like markers. In contrast, laminin increased Lgr5+ stem cells and Paneth cells, and induced crypt-like morphology changes. The transition from a collagen culture to a laminin culture resembled gut development in vivo. The dramatic ECM remodelling was accompanied by a local expression of the laminin receptor ITGA6 in the crypt-forming epithelium. Importantly, deletion of laminin in the adult mouse resulted in a marked reduction of adult intestinal stem cells. Overall, our data support the hypothesis that the formation of intestinal crypts is induced by an increased laminin concentration in the ECM.


Assuntos
Laminina , Células-Tronco , Animais , Camundongos , Colágeno/metabolismo , Matriz Extracelular , Laminina/metabolismo , Laminina/farmacologia , Celulas de Paneth/metabolismo , Intestinos
7.
Cell Stem Cell ; 29(8): 1213-1228.e8, 2022 08 04.
Artigo em Inglês | MEDLINE | ID: mdl-35931031

RESUMO

Intestinal homeostasis is underpinned by LGR5+ve crypt-base columnar stem cells (CBCs), but following injury, dedifferentiation results in the emergence of LGR5-ve regenerative stem cell populations (RSCs), characterized by fetal transcriptional profiles. Neoplasia hijacks regenerative signaling, so we assessed the distribution of CBCs and RSCs in mouse and human intestinal tumors. Using combined molecular-morphological analysis, we demonstrate variable expression of stem cell markers across a range of lesions. The degree of CBC-RSC admixture was associated with both epithelial mutation and microenvironmental signaling disruption and could be mapped across disease molecular subtypes. The CBC-RSC equilibrium was adaptive, with a dynamic response to acute selective pressure, and adaptability was associated with chemoresistance. We propose a fitness landscape model where individual tumors have equilibrated stem cell population distributions along a CBC-RSC phenotypic axis. Cellular plasticity is represented by position shift along this axis and is influenced by cell-intrinsic, extrinsic, and therapeutic selective pressures.


Assuntos
Neoplasias Colorretais , Mucosa Intestinal , Animais , Neoplasias Colorretais/patologia , Homeostase/fisiologia , Humanos , Mucosa Intestinal/metabolismo , Intestinos , Camundongos , Células-Tronco Neoplásicas/patologia , Receptores Acoplados a Proteínas G/metabolismo
8.
Nat Cell Biol ; 23(2): 127-135, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33495632

RESUMO

Ribosomes are multicomponent molecular machines that synthesize all of the proteins of living cells. Most of the genes that encode the protein components of ribosomes are therefore essential. A reduction in gene dosage is often viable albeit deleterious and is associated with human syndromes, which are collectively known as ribosomopathies1-3. The cell biological basis of these pathologies has remained unclear. Here, we model human ribosomopathies in Drosophila and find widespread apoptosis and cellular stress in the resulting animals. This is not caused by insufficient protein synthesis, as reasonably expected. Instead, ribosomal protein deficiency elicits proteotoxic stress, which we suggest is caused by the accumulation of misfolded proteins that overwhelm the protein degradation machinery. We find that dampening the integrated stress response4 or autophagy increases the harm inflicted by ribosomal protein deficiency, suggesting that these activities could be cytoprotective. Inhibition of TOR activity-which decreases ribosomal protein production, slows down protein synthesis and stimulates autophagy5-reduces proteotoxic stress in our ribosomopathy model. Interventions that stimulate autophagy, combined with means of boosting protein quality control, could form the basis of a therapeutic strategy for this class of diseases.


Assuntos
Mutação/genética , Proteínas/toxicidade , Ribossomos/genética , Ribossomos/patologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Alelos , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Drosophila melanogaster/efeitos dos fármacos , Drosophila melanogaster/metabolismo , Células HEK293 , Heterozigoto , Humanos , Discos Imaginais/efeitos dos fármacos , Discos Imaginais/metabolismo , Agregados Proteicos/efeitos dos fármacos , Biossíntese de Proteínas/efeitos dos fármacos , Proteômica , Proteínas Ribossômicas/biossíntese , Transdução de Sinais/efeitos dos fármacos , Serina-Treonina Quinases TOR/metabolismo , Asas de Animais/efeitos dos fármacos , Asas de Animais/metabolismo
9.
Nat Commun ; 12(1): 3464, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34103493

RESUMO

Right-sided (proximal) colorectal cancer (CRC) has a poor prognosis and a distinct mutational profile, characterized by oncogenic BRAF mutations and aberrations in mismatch repair and TGFß signalling. Here, we describe a mouse model of right-sided colon cancer driven by oncogenic BRAF and loss of epithelial TGFß-receptor signalling. The proximal colonic tumours that develop in this model exhibit a foetal-like progenitor phenotype (Ly6a/Sca1+) and, importantly, lack expression of Lgr5 and its associated intestinal stem cell signature. These features are recapitulated in human BRAF-mutant, right-sided CRCs and represent fundamental differences between left- and right-sided disease. Microbial-driven inflammation supports the initiation and progression of these tumours with foetal-like characteristics, consistent with their predilection for the microbe-rich right colon and their antibiotic sensitivity. While MAPK-pathway activating mutations drive this foetal-like signature via ERK-dependent activation of the transcriptional coactivator YAP, the same foetal-like transcriptional programs are also initiated by inflammation in a MAPK-independent manner. Importantly, in both contexts, epithelial TGFß-receptor signalling is instrumental in suppressing the tumorigenic potential of these foetal-like progenitor cells.


Assuntos
Carcinogênese/metabolismo , Neoplasias do Colo/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Transdução de Sinais , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Carcinogênese/patologia , Diferenciação Celular , Sobrevivência Celular , Colo/patologia , Neoplasias do Colo/genética , Células Epiteliais/metabolismo , Feto/patologia , Inflamação/patologia , Estimativa de Kaplan-Meier , Sistema de Sinalização das MAP Quinases , Camundongos Endogâmicos C57BL , Mutação , Prognóstico , Proteínas Proto-Oncogênicas B-raf/genética , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Esferoides Celulares/metabolismo , Esferoides Celulares/patologia , Fatores de Transcrição/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Proteínas de Sinalização YAP
10.
Nat Commun ; 10(1): 1453, 2019 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-30914643

RESUMO

The original version of this Article contained an error in the spelling of the author Miryam Müller, which was incorrectly given as Miryam Müeller. This has now been corrected in both the PDF and HTML versions of the Article.

11.
Nat Commun ; 10(1): 723, 2019 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-30760720

RESUMO

Different thresholds of Wnt signalling are thought to drive stem cell maintenance, regeneration, differentiation and cancer. However, the principle that oncogenic Wnt signalling could be specifically targeted remains controversial. Here we examine the requirement of BCL9/9l, constituents of the Wnt-enhanceosome, for intestinal transformation following loss of the tumour suppressor APC. Although required for Lgr5+ intestinal stem cells and regeneration, Bcl9/9l deletion has no impact upon normal intestinal homeostasis. Loss of BCL9/9l suppressed many features of acute APC loss and subsequent Wnt pathway deregulation in vivo. This resulted in a level of Wnt pathway activation that favoured tumour initiation in the proximal small intestine (SI) and blocked tumour growth in the colon. Furthermore, Bcl9/9l deletion completely abrogated ß-catenin driven intestinal and hepatocellular transformation. We speculate these results support the just-right hypothesis of Wnt-driven tumour formation. Importantly, loss of BCL9/9l is particularly effective at blocking colonic tumourigenesis and mutations that most resemble those that occur in human cancer.


Assuntos
Carcinogênese , Proteínas de Ligação a DNA/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Wnt/metabolismo , Via de Sinalização Wnt , Animais , Transformação Celular Neoplásica , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/genética , Feminino , Deleção de Genes , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Homeostase , Humanos , Mucosa Intestinal/metabolismo , Intestinos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas , Oncogenes , Receptores Acoplados a Proteínas G/metabolismo , Fatores de Transcrição/genética , beta Catenina
12.
Cell Stem Cell ; 22(1): 7-9, 2018 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-29304343

RESUMO

In this issue of Cell Stem Cell, Yui et al. (2018) show how tissue regeneration is driven by changes in the microenvironment. During intestinal regeneration, the epithelium is reprogrammed into a fetal state by an altered extracellular matrix (ECM), which is dependent on YAP/TAZ activation.


Assuntos
Reprogramação Celular , Fosfoproteínas/genética , Proteínas Adaptadoras de Transdução de Sinal/genética , Epitélio , Matriz Extracelular
13.
Dev Cell ; 41(5): 456-458, 2017 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-28586642

RESUMO

The intestinal crypt and its stem cells are dependent on the Wnt pathway. Reporting in Nature, Yan et al. (2017) show that Wnts and Wnt signaling potentiator R-spondins have non-interchangeable roles. Both are necessary for intestinal stem cell (ISC) maintenance, and R-spondins are the limiting factor that defines ISC number.


Assuntos
Trombospondinas , Via de Sinalização Wnt , Humanos , Intestinos , Células-Tronco
14.
Cell Death Differ ; 24(10): 1681-1693, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28622298

RESUMO

Recent studies have suggested increased plasticity of differentiated cells within the intestine to act both as intestinal stem cells (ISCs) and tumour-initiating cells. However, little is known of the processes that regulate this plasticity. Our previous work has shown that activating mutations of Kras or the NF-κB pathway can drive dedifferentiation of intestinal cells lacking Apc. To investigate this process further, we profiled both cells undergoing dedifferentiation in vitro and tumours generated from these cells in vivo by gene expression analysis. Remarkably, no clear differences were observed in the tumours; however, during dedifferentiation in vitro we found a marked upregulation of TGFß signalling, a pathway commonly mutated in colorectal cancer (CRC). Genetic inactivation of TGFß type 1 receptor (Tgfbr1/Alk5) enhanced the ability of KrasG12D/+ mutation to drive dedifferentiation and markedly accelerated tumourigenesis. Mechanistically this is associated with a marked activation of MAPK signalling. Tumourigenesis from differentiated compartments is potently inhibited by MEK inhibition. Taken together, we show that tumours arising in differentiated compartments will be exposed to different suppressive signals, for example, TGFß and blockade of these makes tumourigenesis more efficient from this compartment.


Assuntos
Carcinogênese/genética , Transformação Celular Neoplásica/genética , Regulação Neoplásica da Expressão Gênica/genética , Sistema de Sinalização das MAP Quinases , Fator de Crescimento Transformador beta/metabolismo , Via de Sinalização Wnt/genética , Animais , Proliferação de Células/genética , Genes ras/genética , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação/genética , NF-kappa B/metabolismo , Fator de Crescimento Transformador beta/genética
16.
Cell Rep ; 7(1): 94-103, 2014 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-24685131

RESUMO

Self-renewal is essential for multicellular organisms but carries the risk of somatic mutations that can lead to cancer, which is particularly critical for rapidly renewing tissues in a highly mutagenic environment such as the intestinal epithelium. Using computational modeling and in vivo experimentation, we have analyzed how adenomatous polyposis coli (APC) mutations and ß-catenin aberrations affect the maintenance of mutant cells in colonic crypts. The increasing abundance of APC along the crypt axis forms a gradient of cellular adhesion that causes more proliferative cells to accelerate their movement toward the top of the crypt, where they are shed into the lumen. Thus, the normal crypt can efficiently eliminate ß-catenin mutant cells, whereas APC mutations favor retention. Together, the molecular design of the APC/ß-catenin signaling network integrates cell proliferation and migration dynamics to translate intracellular signal processing and protein gradients along the crypt into intercellular interactions and whole-crypt physiological or pathological behavior.


Assuntos
Polipose Adenomatosa do Colo/metabolismo , Polipose Adenomatosa do Colo/patologia , Polipose Adenomatosa do Colo/genética , Animais , Apoptose/fisiologia , Caderinas/genética , Caderinas/metabolismo , Processos de Crescimento Celular/fisiologia , Colo/metabolismo , Colo/patologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Biológicos , Transdução de Sinais , beta Catenina/genética , beta Catenina/metabolismo
17.
Cell Rep ; 6(6): 1153-1164, 2014 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-24630994

RESUMO

The small G protein family Rac has numerous regulators that integrate extracellular signals into tight spatiotemporal maps of its activity to promote specific cell morphologies and responses. Here, we have generated a mouse strain, Rac-FRET, which ubiquitously expresses the Raichu-Rac biosensor. It enables FRET imaging and quantification of Rac activity in live tissues and primary cells without affecting cell properties and responses. We assessed Rac activity in chemotaxing Rac-FRET neutrophils and found enrichment in leading-edge protrusions and unexpected longitudinal shifts and oscillations during protruding and stalling phases of migration. We monitored Rac activity in normal or disease states of intestinal, liver, mammary, pancreatic, and skin tissue, in response to stimulation or inhibition and upon genetic manipulation of upstream regulators, revealing unexpected insights into Rac signaling during disease development. The Rac-FRET strain is a resource that promises to fundamentally advance our understanding of Rac-dependent responses in primary cells and native environments.


Assuntos
Neutrófilos/enzimologia , Proteínas rac de Ligação ao GTP/metabolismo , Animais , Ativação Enzimática , Transferência Ressonante de Energia de Fluorescência/métodos , Camundongos , Neutrófilos/citologia , Transdução de Sinais , Análise Espaço-Temporal , Proteínas rac de Ligação ao GTP/química
18.
Methods Mol Biol ; 1012: 237-48, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24006069

RESUMO

Within the intestinal epithelium, c-Myc has been characterized as a target of ß-catenin-TCF signalling (He et al., Science 281:1509-1512, 1998). Given the most commonly mutated tumor suppressor gene within colorectal cancer (CRC) is the APC (Adenomatous Polyposis Coli) gene, a negative regulator of ß-catenin-TCF signalling (Korinek et al., Science 275:1784-1787, 1997), loss of APC leads to Myc deregulation in the vast majority of CRC. This probably explains the numerous studies investigating c-Myc function within the intestinal epithelium. These have shown that c-Myc inhibition or deletion in the adult intestine results in proliferative defects (Muncan et al., Mol Cell Biol 26:8418-8426, 2006; Soucek et al., Nature 455:679-683, 2008). Importantly, intestinal enterocytes are able to survive in the absence of c-Myc which has allowed us (and others) to test the role of c-Myc in intestinal regeneration and tumorigenesis. Remarkably c-Myc deletion suppresses all the phenotypes of the Apc tumor suppressor gene loss and stops intestinal regeneration (Ashton et al., Dev Cell 19:259-269, 2010; Sansom et al., Oncogene 29:2585-2590, 2007). This suggests a clear therapeutic rationale for targeting c-Myc in CRC. Moreover haploinsufficiency for c-Myc in this tissue also reduces intestinal tumorigenesis (Athineos and Sansom, Oncogene 29:2585-2590, 2010; Yekkala and Baudino, Mol Cancer Res 5:1296-1303, 2007), and overexpression of c-Myc affects tissue homeostasis (Finch et al., Mol Cell Biol 29:5306-5315, 2009; Murphy et al., Cancer Cell 14:447-457, 2008). In this chapter we will provide an overview of our current laboratory protocols to characterize c-Myc function in intestinal homeostasis, regeneration, and tumorigenesis in vivo and in vitro.


Assuntos
Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Homeostase/genética , Mucosa Intestinal/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Regeneração/genética , Técnicas de Cultura de Células , Enterócitos/metabolismo , Enterócitos/patologia , Humanos , Mucosa Intestinal/patologia , Intestinos/patologia , Esferoides Celulares , Técnicas de Cultura de Tecidos , Células Tumorais Cultivadas
19.
Cell Stem Cell ; 12(6): 761-73, 2013 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-23665120

RESUMO

The Adenomatous Polyposis Coli (APC) gene is mutated in the majority of colorectal cancers (CRCs). Loss of APC leads to constitutively active WNT signaling, hyperproliferation, and tumorigenesis. Identification of pathways that facilitate tumorigenesis after APC loss is important for therapeutic development. Here, we show that RAC1 is a critical mediator of tumorigenesis after APC loss. We find that RAC1 is required for expansion of the LGR5 intestinal stem cell (ISC) signature, progenitor hyperproliferation, and transformation. Mechanistically, RAC1-driven ROS and NF-κB signaling mediate these processes. Together, these data highlight that ROS production and NF-κB activation triggered by RAC1 are critical events in CRC initiation.


Assuntos
Neoplasias Colorretais/patologia , Intestino Delgado/citologia , NF-kappa B/metabolismo , Neuropeptídeos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células-Tronco/citologia , Proteínas Wnt/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Animais , Proliferação de Células , Neoplasias Colorretais/metabolismo , Intestino Delgado/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais , Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA