Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
iScience ; 27(5): 109718, 2024 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-38706869

RESUMO

Cell competition plays an instrumental role in quality control during tissue development and homeostasis. Nevertheless, cancer cells can exploit this process for their own proliferative advantage. In our study, we generated mixed murine organoids and microtissues to explore the impact of cell competition on liver metastasis. Unlike competition at the primary site, the initial effect on liver progenitor cells does not involve the induction of apoptosis. Instead, metastatic competition manifests as a multistage process. Initially, liver progenitors undergo compaction, which is followed by cell-cycle arrest, ultimately forcing differentiation. Subsequently, the newly differentiated liver cells exhibit reduced cellular fitness, rendering them more susceptible to outcompetition by intestinal cancer cells. Notably, cancer cells leverage different interactions with different epithelial populations in the liver, using them as scaffolds to facilitate their growth. Consequently, tissue-specific mechanisms of cell competition are fundamental in driving metastatic intestinal cancer.

2.
J Exp Clin Cancer Res ; 42(1): 56, 2023 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-36869386

RESUMO

BACKGROUND: Colorectal cancer (CRC) can be divided into four consensus molecular subtypes (CMS), each with distinct biological features. CMS4 is associated with epithelial-mesenchymal transition and stromal infiltration (Guinney et al., Nat Med 21:1350-6, 2015; Linnekamp et al., Cell Death Differ 25:616-33, 2018), whereas clinically it is characterized by lower responses to adjuvant therapy, higher incidence of metastatic spreading and hence dismal prognosis (Buikhuisen et al., Oncogenesis 9:66, 2020). METHODS: To understand the biology of the mesenchymal subtype and unveil specific vulnerabilities, a large CRISPR-Cas9 drop-out screen was performed on 14 subtyped CRC cell lines to uncover essential kinases in all CMSs. Dependency of CMS4 cells on p21-activated kinase 2 (PAK2) was validated in independent 2D and 3D in vitro cultures and in vivo models assessing primary and metastatic outgrowth in liver and peritoneum. TIRF microscopy was used to uncover actin cytoskeleton dynamics and focal adhesion localization upon PAK2 loss. Subsequent functional assays were performed to determine altered growth and invasion patterns. RESULTS: PAK2 was identified as a key kinase uniquely required for growth of the mesenchymal subtype CMS4, both in vitro and in vivo. PAK2 plays an important role in cellular attachment and cytoskeletal rearrangements (Coniglio et al., Mol Cell Biol 28:4162-72, 2008; Grebenova et al., Sci Rep 9:17171, 2019). In agreement, deletion or inhibition of PAK2 impaired actin cytoskeleton dynamics in CMS4 cells and, as a consequence, significantly reduced invasive capacity, while it was dispensable for CMS2 cells. Clinical relevance of these findings was supported by the observation that deletion of PAK2 from CMS4 cells prevented metastatic spreading in vivo. Moreover, growth in a model for peritoneal metastasis was hampered when CMS4 tumor cells were deficient for PAK2. CONCLUSION: Our data reveal a unique dependency of mesenchymal CRC and provide a rationale for PAK2 inhibition to target this aggressive subgroup of colorectal cancer.


Assuntos
Neoplasias Colorretais , Sarcoma , Humanos , Citoesqueleto de Actina , Carcinogênese , Linhagem Celular
3.
Nat Commun ; 13(1): 4492, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35918345

RESUMO

The small intestine is a rapidly proliferating organ that is maintained by a small population of Lgr5-expressing intestinal stem cells (ISCs). However, several Lgr5-negative ISC populations have been identified, and this remarkable plasticity allows the intestine to rapidly respond to both the local environment and to damage. However, the mediators of such plasticity are still largely unknown. Using intestinal organoids and mouse models, we show that upon ribosome impairment (driven by Rptor deletion, amino acid starvation, or low dose cyclohexamide treatment) ISCs gain an Lgr5-negative, fetal-like identity. This is accompanied by a rewiring of metabolism. Our findings suggest that the ribosome can act as a sensor of nutrient availability, allowing ISCs to respond to the local nutrient environment. Mechanistically, we show that this phenotype requires the activation of ZAKɑ, which in turn activates YAP, via SRC. Together, our data reveals a central role for ribosome dynamics in intestinal stem cells, and identify the activation of ZAKɑ as a critical mediator of stem cell identity.


Assuntos
Mucosa Intestinal , Células-Tronco , Animais , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Intestinos , Camundongos , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Ribossomos/metabolismo , Células-Tronco/metabolismo
4.
STAR Protoc ; 2(4): 100997, 2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34917977

RESUMO

Cell competition is a mechanism of interaction that dictates cell selection based on differences in cellular fitness. We designed a protocol to generate mixed murine organoids and enteroid monolayers used to study such complex cellular interactions in a mammalian system. This protocol is dedicated to follow the behavior of different cell populations over time, using (time-lapse) microscopy or transcriptome/proteome analysis. For complete details on the use and execution of this protocol, please refer to Krotenberg Garcia et al. (2021).


Assuntos
Comunicação Celular/fisiologia , Organoides/citologia , Técnicas de Cultura de Tecidos/métodos , Animais , Técnicas de Cultura de Células , Células Cultivadas , Camundongos
5.
Cell Rep ; 36(1): 109307, 2021 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-34233177

RESUMO

Competitive cell interactions play a crucial role in quality control during development and homeostasis. Here, we show that cancer cells use such interactions to actively eliminate wild-type intestine cells in enteroid monolayers and organoids. This apoptosis-dependent process boosts proliferation of intestinal cancer cells. The remaining wild-type population activates markers of primitive epithelia and transits to a fetal-like state. Prevention of this cell-state transition avoids elimination of wild-type cells and, importantly, limits the proliferation of cancer cells. Jun N-terminal kinase (JNK) signaling is activated in competing cells and is required for cell-state change and elimination of wild-type cells. Thus, cell competition drives growth of cancer cells by active out-competition of wild-type cells through forced cell death and cell-state change in a JNK-dependent manner.


Assuntos
Carcinogênese/patologia , Intestinos/patologia , Organoides/patologia , Animais , Apoptose , Carcinogênese/metabolismo , Competição entre as Células , Linhagem Celular Tumoral , Feto/patologia , Sistema de Sinalização das MAP Quinases , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Organoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Células-Tronco/metabolismo
6.
Cell Rep ; 32(3): 107937, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32698002

RESUMO

Calorie restriction (CR) extends lifespan through several intracellular mechanisms, including increased DNA repair, leading to fewer DNA mutations that cause age-related pathologies. However, it remains unknown how CR acts on mutation retention at the tissue level. Here, we use Cre-mediated DNA recombination of the confetti reporter as proxy for neutral mutations and follow these mutations by intravital microscopy to identify how CR affects retention of mutations in the intestine. We find that CR leads to increased numbers of functional Lgr5+ stem cells that compete for niche occupancy, resulting in slower but stronger stem cell competition. Consequently, stem cells carrying neutral or Apc mutations encounter more wild-type competitors, thus increasing the chance that they get displaced from the niche to get lost over time. Thus, our data show that CR not only affects the acquisition of mutations but also leads to lower retention of mutations in the intestine.


Assuntos
Restrição Calórica , Competição entre as Células , Intestinos/citologia , Mutação/genética , Células-Tronco/citologia , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Contagem de Células , Linhagem da Célula , Feminino , Microscopia Intravital , Masculino , Camundongos Endogâmicos C57BL
7.
Cell Stem Cell ; 26(4): 569-578.e7, 2020 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-32169167

RESUMO

Colorectal cancer stem cells (CSCs) express Lgr5 and display extensive stem cell-like multipotency and self-renewal and are thought to seed metastatic disease. Here, we used a mouse model of colorectal cancer (CRC) and human tumor xenografts to investigate the cell of origin of metastases. We found that most disseminated CRC cells in circulation were Lgr5- and formed distant metastases in which Lgr5+ CSCs appeared. This plasticity occurred independently of stemness-inducing microenvironmental factors and was indispensable for outgrowth, but not establishment, of metastases. Together, these findings show that most colorectal cancer metastases are seeded by Lgr5- cells, which display intrinsic capacity to become CSCs in a niche-independent manner and can restore epithelial hierarchies in metastatic tumors.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Biomarcadores Tumorais , Humanos , Células-Tronco Neoplásicas , Receptores Acoplados a Proteínas G
8.
Nat Protoc ; 13(2): 235-247, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29300390

RESUMO

Most currently available colorectal cancer (CRC) mouse models are not suitable for studying progression toward the metastatic stage. Recently, establishment of tumor organoid lines, either from murine CRC models or patients, and the possibility of engineering them with genome-editing technologies, have provided a large collection of tumor material faithfully recapitulating phenotypic and genetic heterogeneity of native tumors. To study tumor progression in the natural in vivo environment, we developed an orthotopic approach based on transplantation of CRC organoids into the cecal epithelium. The 20-min procedure is described in detail here and enables growth of transplanted organoids into a single tumor mass within the intestinal tract. Due to long latency, tumor cells are capable of spreading through the blood circulation and forming metastases at distant sites. This method is designed to generate tumors suitable for studying CRC progression, thereby providing the opportunity to visualize tumor cell dynamics in vivo in real time by intravital microscopy.


Assuntos
Neoplasias Colorretais/fisiopatologia , Edição de Genes/métodos , Organoides/transplante , Animais , Sistemas CRISPR-Cas/genética , Transformação Celular Neoplásica , Neoplasias do Colo , Colonoscopia/métodos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Hepáticas , Camundongos , Camundongos Endogâmicos NOD , Organoides/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Dev Biol ; 428(2): 328-337, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28473106

RESUMO

Homeostasis of tissues is tightly regulated at the cellular, tissue and organismal level. Interestingly, tumor cells have found ways to hijack many of these physiological processes at all the different levels. Here we review how intravital microscopy techniques have provided new insights into our understanding of tissue homeostasis and cancer progression. In addition, we highlight the different strategies that tumor cells have adopted to use these physiological processes for their own benefit. We describe how visualization of these dynamic processes in living mice has broadened to our view on cancer initiation and progression.


Assuntos
Neoplasias/fisiopatologia , Células-Tronco Adultas/patologia , Células-Tronco Adultas/fisiologia , Animais , Comunicação Celular/fisiologia , Movimento Celular/fisiologia , Progressão da Doença , Humanos , Inflamação/patologia , Inflamação/fisiopatologia , Microscopia Intravital/métodos , Neoplasias/etiologia , Neoplasias/patologia , Células-Tronco Neoplásicas/patologia , Células-Tronco Neoplásicas/fisiologia , Neovascularização Patológica , Nicho de Células-Tronco/fisiologia , Microambiente Tumoral/fisiologia , Cicatrização/fisiologia
10.
Proc Natl Acad Sci U S A ; 114(12): E2357-E2364, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28270604

RESUMO

In the adenoma-carcinoma sequence, it is proposed that intestinal polyps evolve through a set of defined mutations toward metastatic colorectal cancer (CRC). Here, we dissect this adenoma-carcinoma sequence in vivo by using an orthotopic organoid transplantation model of human colon organoids engineered to harbor different CRC mutation combinations. We demonstrate that sequential accumulation of oncogenic mutations in Wnt, EGFR, P53, and TGF-ß signaling pathways facilitates efficient tumor growth, migration, and metastatic colonization. We show that reconstitution of specific niche signals can restore metastatic growth potential of tumor cells lacking one of the oncogenic mutations. Our findings imply that the ability to metastasize-i.e., to colonize distant sites-is the direct consequence of the loss of dependency on specific niche signals.


Assuntos
Neoplasias Colorretais/genética , Organoides/transplante , Adulto , Animais , Movimento Celular , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Neoplasias Colorretais/fisiopatologia , Modelos Animais de Doenças , Receptores ErbB/genética , Receptores ErbB/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Engenharia Genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Pessoa de Meia-Idade , Mutação , Metástase Neoplásica/genética , Processos Neoplásicos , Organoides/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
11.
Curr Biol ; 26(4): 428-38, 2016 Feb 22.
Artigo em Inglês | MEDLINE | ID: mdl-26853366

RESUMO

Tumor-host interactions play an increasingly recognized role in modulating tumor growth. Thus, understanding the nature and impact of this complex bidirectional communication is key to identifying successful anti-cancer strategies. It has been proposed that tumor cells compete with and kill neighboring host tissue to clear space that they can expand into; however, this has not been demonstrated experimentally. Here we use the adult fly intestine to investigate the existence and characterize the role of competitive tumor-host interactions. We show that APC(-/-)-driven intestinal adenomas compete with and kill surrounding cells, causing host tissue attrition. Importantly, we demonstrate that preventing cell competition, by expressing apoptosis inhibitors, restores host tissue growth and contains adenoma expansion, indicating that cell competition is essential for tumor growth. We further show that JNK signaling is activated inside the tumor and in nearby tissue and is required for both tumor growth and cell competition. Lastly, we find that APC(-/-) cells display higher Yorkie (YAP) activity than host cells and that this promotes tumor growth, in part via cell competition. Crucially, we find that relative, rather than absolute, Hippo activity determines adenoma growth. Overall, our data indicate that the intrinsic over-proliferative capacity of APC(-/-) cells is not uncontrolled and can be constrained by host tissues if cell competition is inhibited, suggesting novel possible therapeutic approaches.


Assuntos
Adenoma/etiologia , Carcinogênese , Transformação Celular Neoplásica , Drosophila melanogaster/crescimento & desenvolvimento , Neoplasias Intestinais/etiologia , Adenoma/fisiopatologia , Animais , Proliferação de Células , Modelos Animais de Doenças , Drosophila melanogaster/citologia , Humanos , Neoplasias Intestinais/fisiopatologia , Transdução de Sinais
12.
Dev Cell ; 34(3): 297-309, 2015 Aug 10.
Artigo em Inglês | MEDLINE | ID: mdl-26212135

RESUMO

Throughout their lifetime, cells may suffer insults that reduce their fitness and disrupt their function, and it is unclear how these potentially harmful cells are managed in adult tissues. We address this question using the adult Drosophila posterior midgut as a model of homeostatic tissue and ribosomal Minute mutations to reduce fitness in groups of cells. We take a quantitative approach combining lineage tracing and biophysical modeling and address how cell competition affects stem cell and tissue population dynamics. We show that healthy cells induce clonal extinction in weak tissues, targeting both stem and differentiated cells for elimination. We also find that competition induces stem cell proliferation and self-renewal in healthy tissue, promoting selective advantage and tissue colonization. Finally, we show that winner cell proliferation is fueled by the JAK-STAT ligand Unpaired-3, produced by Minute(-/+) cells in response to chronic JNK stress signaling.


Assuntos
Células-Tronco Adultas/citologia , Proteínas de Drosophila/metabolismo , Janus Quinases/metabolismo , Sistema de Sinalização das MAP Quinases/fisiologia , Fatores de Transcrição STAT/metabolismo , Fatores de Transcrição/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem da Célula , Proliferação de Células , Drosophila melanogaster/citologia , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Ribossomos/genética
13.
Dev Cell ; 23(4): 745-55, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-23079597

RESUMO

Maintenance of chromosomal stability depends on error-free chromosome segregation. The pseudokinase BUBR1 is essential for this, because it is a core component of the mitotic checkpoint and is required for formation of stable kinetochore-microtubule attachments. We have identified a conserved and highly phosphorylated domain (KARD) in BUBR1 that is crucial for formation of kinetochore-microtubule attachments. Deletion of this domain or prevention of its phosphorylation abolishes formation of kinetochore microtubules, which can be reverted by inhibiting Aurora B activity. Phosphorylation of KARD by PLK1 promotes direct interaction of BUBR1 with the PP2A-B56α phosphatase that counters excessive Aurora B activity at kinetochores. As a result, removal of BUBR1 from mitotic cells or inhibition of PLK1 reduces PP2A-B56α kinetochore binding and elevates phosphorylation of Aurora B substrates on the outer kinetochore. We propose that PLK1 and BUBR1 cooperate to stabilize kinetochore-microtubule interactions by regulating PP2A-B56α-mediated dephosphorylation of Aurora B substrates at the kinetochore-microtubule interface.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cinetocoros/metabolismo , Microtúbulos/metabolismo , Proteína Fosfatase 2/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Aurora Quinase B , Aurora Quinases , Células HEK293 , Células HeLa , Humanos , Fosforilação , Células Tumorais Cultivadas , Quinase 1 Polo-Like
14.
Dev Cell ; 22(6): 1321-9, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22698286

RESUMO

Chromosomal stability is safeguarded by a mitotic checkpoint, of which BUB1 and Mad3/BUBR1 are core components. These paralogs have similar, but not identical, domain organization. We show that Mad3/BUBR1 and BUB1 paralogous pairs arose by nine independent gene duplications throughout evolution, followed by parallel subfunctionalization in which preservation of the ancestral, amino-terminal KEN box or kinase domain was mutually exclusive. In one exception, vertebrate BUBR1-defined by the KEN box-preserved the kinase domain but allowed nonconserved degeneration of catalytic motifs. Although BUBR1 evolved to a typical pseudokinase in some vertebrates, it retained the catalytic triad in humans. However, we show that putative catalysis by human BUBR1 is dispensable for error-free chromosome segregation. Instead, residues that interact with ATP in conventional kinases are essential for conformational stability in BUBR1. We propose that parallel evolution of BUBR1 orthologs rendered its kinase function dispensable in vertebrates, producing an unusual, triad-containing pseudokinase.


Assuntos
Pontos de Checagem da Fase M do Ciclo Celular , Proteínas Serina-Treonina Quinases/metabolismo , Sequência de Aminoácidos , Animais , Sequência de Bases , Evolução Biológica , Segregação de Cromossomos , Duplicação Gênica , Humanos , Lagartos , Dados de Sequência Molecular , Mutação , Conformação Proteica , Proteínas Serina-Treonina Quinases/genética , Alinhamento de Sequência , Proteínas de Peixe-Zebra/genética
15.
Cancer Res ; 70(12): 4891-900, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20516114

RESUMO

Genetic mutations in the mitotic regulatory kinase BUBR1 are associated with the cancer-susceptible disorder mosaic variegated aneuploidy (MVA). In patients with biallelic mutations, a missense mutation pairs with a truncating mutation. Here, we show that cell lines derived from MVA patients with biallelic mutations have an impaired mitotic checkpoint, chromosome alignment defects, and low overall BUBR1 abundance. Ectopic expression of BUBR1 restored mitotic checkpoint activity, proving that BUBR1 dysfunction causes chromosome segregation errors in the patients. Combined analysis of patient cells and functional protein replacement shows that all MVA mutations fall in two distinct classes: those that impose specific defects in checkpoint activity or microtubule attachment and those that lower BUBR1 protein abundance. Low protein abundance is the direct result of the absence of transcripts from truncating mutants combined with high protein turnover of missense mutants. In this group of missense mutants, the amino acid change consistently occurs in or near the BUBR1 kinase domain. Our findings provide a molecular explanation for chromosomal instability in patients with biallelic genetic mutations in BUBR1.


Assuntos
Anormalidades Múltiplas/genética , Aneuploidia , Segregação de Cromossomos/genética , Predisposição Genética para Doença , Mutação/genética , Neoplasias/genética , Proteínas Serina-Treonina Quinases/genética , Northern Blotting , Citometria de Fluxo , Genes cdc/fisiologia , Células HeLa , Humanos , Immunoblotting , Mosaicismo , Neoplasias/patologia , Plasmídeos , Proteínas Serina-Treonina Quinases/metabolismo , Fuso Acromático/patologia , Síndrome , Transfecção , Células Tumorais Cultivadas
16.
PLoS Biol ; 6(9): e224, 2008 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-18798690

RESUMO

Centrosomes comprise a pair of centrioles surrounded by an amorphous pericentriolar material (PCM). Here, we have performed a microscopy-based genome-wide RNA interference (RNAi) screen in Drosophila cells to identify proteins required for centriole duplication and mitotic PCM recruitment. We analysed 92% of the Drosophila genome (13,059 genes) and identified 32 genes involved in centrosome function. An extensive series of secondary screens classified these genes into four categories: (1) nine are required for centriole duplication, (2) 11 are required for centrosome maturation, (3) nine are required for both functions, and (4) three genes regulate centrosome separation. These 32 hits include several new centrosomal components, some of which have human homologs. In addition, we find that the individual depletion of only two proteins, Polo and Centrosomin (Cnn) can completely block centrosome maturation. Cnn is phosphorylated during mitosis in a Polo-dependent manner, suggesting that the Polo-dependent phosphorylation of Cnn initiates centrosome maturation in flies.


Assuntos
Centríolos/metabolismo , Centrossomo/metabolismo , Drosophila melanogaster/genética , Genoma , Interferência de RNA , Sequência de Aminoácidos , Animais , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Ciclo Celular/fisiologia , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Centrossomo/ultraestrutura , Proteínas de Drosophila/genética , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/citologia , Drosophila melanogaster/metabolismo , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Dados de Sequência Molecular , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Fuso Acromático/genética , Fuso Acromático/metabolismo
17.
Biochim Biophys Acta ; 1786(1): 24-31, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18472014

RESUMO

Aneuploidy, an abnormal number of chromosomes, is a trait shared by most solid tumors. Chromosomal instability (CIN) manifested as aneuploidy might promote tumorigenesis and cause increased resistance to anti-cancer therapies. The mitotic checkpoint or spindle assembly checkpoint is a major signaling pathway involved in the prevention of CIN. We review current knowledge on the contribution of misregulation of mitotic checkpoint proteins to tumor formation and will address to what extent this contribution is due to chromosome segregation errors directly. We propose that both checkpoint and non-checkpoint functions of these proteins contribute to the wide array of oncogenic phenotypes seen upon their misregulation.


Assuntos
Aneuploidia , Mitose/fisiologia , Neoplasias/etiologia , Animais , Proteínas Cdc20 , Proteínas de Ciclo Celular/fisiologia , Sobrevivência Celular , Instabilidade Cromossômica , Segregação de Cromossomos/fisiologia , Humanos , Proteínas Mad2 , Camundongos , Modelos Animais , Proteínas Nucleares/fisiologia , Prometáfase/fisiologia , Proteínas Serina-Treonina Quinases , Proteínas de Saccharomyces cerevisiae/fisiologia
18.
Gastroenterology ; 132(2): 628-32, 2007 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17320548

RESUMO

BACKGROUND & AIMS: In colorectal cancer, activating mutations in the Wnt pathway transform epithelial cells through the inappropriate expression of a TCF4 target gene program, which is physiologically expressed in intestinal crypts. METHODS: We have now performed an exhaustive array-based analysis of this target gene program in colorectal cancer cell lines carrying an inducible block of the Wnt cascade. Independently, differential gene-expression profiles of human adenomas and adenocarcinomas vs normal colonic epithelium were obtained. RESULTS: Expression analyses of approximately 80 genes common between these data sets were performed in a murine adenoma model. The combined data sets describe a core target gene program, the intestinal Wnt/TCF signature gene set, which is responsible for the transformation of human intestinal epithelial cells. CONCLUSIONS: The genes were invariably expressed in adenomas, yet could be subdivided into 3 modules, based on expression in distinct crypt compartments. A module of 17 genes was specifically expressed at the position of the crypt stem cell.


Assuntos
Transformação Celular Neoplásica/metabolismo , Regulação Neoplásica da Expressão Gênica , Mucosa Intestinal/metabolismo , Transdução de Sinais , Fatores de Transcrição TCF/metabolismo , Proteínas Wnt/metabolismo , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenoma/genética , Adenoma/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Perfilação da Expressão Gênica , Humanos , Mucosa Intestinal/patologia , Camundongos , Análise de Sequência com Séries de Oligonucleotídeos , Celulas de Paneth/metabolismo , RNA Mensageiro/metabolismo , Transdução de Sinais/genética , Fator 1 de Transcrição de Linfócitos T/metabolismo , Fatores de Transcrição TCF/genética , Fatores de Tempo , Proteína 2 Semelhante ao Fator 7 de Transcrição , Transfecção , Proteínas Wnt/genética , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...