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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Br J Cancer ; 113(1): 1-5, 2015 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-26110974

RESUMO

Colorectal cancer (CRC) is one of the most common cancers in the western world and is characterised by deregulation of the Wnt signalling pathway. Mutation of the adenomatous polyposis coli (APC) tumour suppressor gene, which encodes a protein that negatively regulates this pathway, occurs in almost 80% of CRC cases. The progression of this cancer from an early adenoma to carcinoma is accompanied by a well-characterised set of mutations including KRAS, SMAD4 and TP53. Using elegant genetic models the current paradigm is that the intestinal stem cell is the origin of CRC. However, human histology and recent studies, showing marked plasticity within the intestinal epithelium, may point to other cells of origin. Here we will review these latest studies and place these in context to provide an up-to-date view of the cell of origin of CRC.


Assuntos
Neoplasias Colorretais/patologia , Predisposição Genética para Doença , Intestinos/patologia , Células-Tronco/patologia , Neoplasias Colorretais/genética , Humanos , Mutação
2.
Br J Cancer ; 108(5): 997-1003, 2013 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-23385734

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive malignancy associated with <5% 5-year survival, in which standard chemotherapeutics have limited benefit. The disease is associated with significant intra- and peritumoral inflammation and failure of protective immunosurveillance. Indeed, inflammatory signals are implicated in both tumour initiation and tumour progression. The major pathways regulating PDAC-associated inflammation are now being explored. Activation of leukocytes, and upregulation of cytokine and chemokine signalling pathways, both have been shown to modulate PDAC progression. Therefore, targeting inflammatory pathways may be of benefit as part of a multi-target approach to PDAC therapy. This review explores the pathways known to modulate inflammation at different stages of tumour development, drawing conclusions on their potential as therapeutic targets in PDAC.


Assuntos
Carcinoma Ductal Pancreático/imunologia , Carcinoma Ductal Pancreático/terapia , Vigilância Imunológica , Inflamação/terapia , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/terapia , Animais , Carcinoma Ductal Pancreático/patologia , Quimiocinas/metabolismo , Citocinas/metabolismo , Humanos , Macrófagos/imunologia , Terapia de Alvo Molecular , Mutação , Metástase Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Prognóstico , Transdução de Sinais , Evasão Tumoral , Microambiente Tumoral/imunologia
3.
Br J Cancer ; 107(9): 1554-63, 2012 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-23093296

RESUMO

BACKGROUND: Cell line models suggest that activation of NFκB is associated with progression of prostate cancer. This pathway may be a therapeutic target if these observations translate to clinical specimens. METHODS: Immunohistochemistry measured NFκBp65 (p65), NFκBp65 nuclear localisation signal (NLS), NFκBp65 phosphorylated at ser 276 (p65(ser276)), NFκBp65 phosphorylated at ser 536 (p65(ser536)), IκBα phosphorylated at ser 32/36 (pIκBα(ser32/36)) and MMP-9 protein expression in 61 matched hormone naive prostate cancer (HNPC) and castrate-resistant prostate cancer (CRPC) tumours. Animal and cell models were used to investigate the role of NFκB inhibition in prostate carcinogenesis. RESULTS: In HNPC tumours, NLS expression significantly associated with a shorter time to disease recurrence and disease-specific death. In CRPC tumours p65, pIκBα(ser32/36) and MMP-9 expression significantly associated with shorter time to death from disease recurrence and shorter disease-specific death. MMP-9 and pIκBα(ser32/36) expression significantly associated with metastases at recurrence and were independent of Gleason sum and prostate-specific antigen at recurrence. Expression of phosphorylated Akt was associated with increased p65 activation in mouse models and inhibition of NFκB in LNCaP cells significantly reduced cellular proliferation and induced apoptosis. CONCLUSION: These results provide further evidence that the NFκB pathway could be exploited as a target for CRPC.


Assuntos
NF-kappa B/metabolismo , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Idoso , Animais , Biomarcadores Tumorais/biossíntese , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Estudos de Coortes , Modelos Animais de Doenças , Progressão da Doença , Humanos , Proteínas I-kappa B/metabolismo , Imuno-Histoquímica , Estimativa de Kaplan-Meier , Masculino , Metaloproteinase 9 da Matriz/biossíntese , Camundongos , Inibidor de NF-kappaB alfa , NF-kappa B/biossíntese , NF-kappa B/genética , Neoplasias Hormônio-Dependentes/genética , Neoplasias Hormônio-Dependentes/metabolismo , Neoplasias Hormônio-Dependentes/patologia , Fosforilação , Neoplasias da Próstata/genética , Transdução de Sinais , Fator de Transcrição RelA/metabolismo , Regulação para Cima
4.
Nat Commun ; 12(1): 56, 2021 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-33397922

RESUMO

RAC1 activity is critical for intestinal homeostasis, and is required for hyperproliferation driven by loss of the tumour suppressor gene Apc in the murine intestine. To avoid the impact of direct targeting upon homeostasis, we reasoned that indirect targeting of RAC1 via RAC-GEFs might be effective. Transcriptional profiling of Apc deficient intestinal tissue identified Vav3 and Tiam1 as key targets. Deletion of these indicated that while TIAM1 deficiency could suppress Apc-driven hyperproliferation, it had no impact upon tumourigenesis, while VAV3 deficiency had no effect. Intriguingly, deletion of either gene resulted in upregulation of Vav2, with subsequent targeting of all three (Vav2-/- Vav3-/- Tiam1-/-), profoundly suppressing hyperproliferation, tumourigenesis and RAC1 activity, without impacting normal homeostasis. Critically, the observed RAC-GEF dependency was negated by oncogenic KRAS mutation. Together, these data demonstrate that while targeting RAC-GEF molecules may have therapeutic impact at early stages, this benefit may be lost in late stage disease.


Assuntos
Carcinogênese/metabolismo , Carcinogênese/patologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Intestinos/patologia , Transdução de Sinais , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Carcinogênese/genética , Homeostase , Intestinos/ultraestrutura , Camundongos Knockout , Mutação/genética , Especificidade de Órgãos , Fenótipo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/genética , Proteína 1 Indutora de Invasão e Metástase de Linfoma de Células T/metabolismo , Regulação para Cima , Via de Sinalização Wnt
5.
Nat Commun ; 11(1): 445, 2020 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-31974352

RESUMO

The number of patients diagnosed with chronic bile duct disease is increasing and in most cases these diseases result in chronic ductular scarring, necessitating liver transplantation. The formation of ductular scaring affects liver function; however, scar-generating portal fibroblasts also provide important instructive signals to promote the proliferation and differentiation of biliary epithelial cells. Therefore, understanding whether we can reduce scar formation while maintaining a pro-regenerative microenvironment will be essential in developing treatments for biliary disease. Here, we describe how regenerating biliary epithelial cells express Wnt-Planar Cell Polarity signalling components following bile duct injury and promote the formation of ductular scars by upregulating pro-fibrogenic cytokines and positively regulating collagen-deposition. Inhibiting the production of Wnt-ligands reduces the amount of scar formed around the bile duct, without reducing the development of the pro-regenerative microenvironment required for ductular regeneration, demonstrating that scarring and regeneration can be uncoupled in adult biliary disease and regeneration.


Assuntos
Doenças dos Ductos Biliares/patologia , Colangite Esclerosante/patologia , Cicatriz/patologia , Via de Sinalização Wnt , Animais , Proteína Axina/genética , Proteína Axina/metabolismo , Doenças dos Ductos Biliares/induzido quimicamente , Doenças dos Ductos Biliares/metabolismo , Ductos Biliares/citologia , Polaridade Celular , Colangite Esclerosante/metabolismo , Cicatriz/metabolismo , Modelos Animais de Doenças , Células Epiteliais , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , MAP Quinase Quinase 4/metabolismo , Masculino , Camundongos Transgênicos , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Piridinas/toxicidade , Via de Sinalização Wnt/efeitos dos fármacos , Proteína Wnt-5a/metabolismo
6.
Oncogene ; 26(43): 6349-55, 2007 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-17452983

RESUMO

The p53-binding protein 53BP1 has been implicated in the DNA damage response and genomic instability. Previous reports have highlighted these roles in vivo in haematopoietic lineages. To investigate the importance of 53BP1 to the DNA damage response in epithelial cells in vivo, we have investigated the role of 53BP1 in mediating apoptosis and proliferation within the murine small intestine following gamma-irradiation. 53BP1 deficiency does not affect the immediate response to gamma-irradiation with normal levels of apoptosis, proliferation and p53 and p21 accumulation. However, 48 h post-gamma-irradiation there was a significant accumulation of cells with much larger nuclei marked by p53 and p21 accumulation. These data reflect increases in polyploidy observed 53BP1-/- deficient fibroblasts following gamma-irradiation. At 72 h post-irradiation both the 4N and 8N populations were significantly increased in 53BP1-/- MEFS. Taken together, these results show that following in vivo exposure to gamma-irradiation, 53BP1 is dispensable for signalling apoptosis and cell-cycle arrest in the intestinal epithelium. However, it is important for prevention of genomic instability within this epithelial cell population.


Assuntos
Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Enterócitos/metabolismo , Mucosa Intestinal/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fosfoproteínas/deficiência , Fosfoproteínas/metabolismo , Poliploidia , Animais , Apoptose/efeitos da radiação , Células Cultivadas , Proteínas Cromossômicas não Histona , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Proteínas de Ligação a DNA , Enterócitos/efeitos da radiação , Intestinos/efeitos da radiação , Peptídeos e Proteínas de Sinalização Intracelular/genética , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , Radiação Ionizante , Proteína Supressora de Tumor p53/metabolismo , Proteína 1 de Ligação à Proteína Supressora de Tumor p53
7.
Oncogene ; 37(8): 982-992, 2018 02 22.
Artigo em Inglês | MEDLINE | ID: mdl-29106388

RESUMO

NUAK1 is a member of the AMPK-related family of kinases. Recent evidence suggests that NUAK1 is an important regulator of cell adhesion and migration, cellular and organismal metabolism, and regulation of TAU stability. As such, NUAK1 may play key roles in multiple diseases ranging from neurodegeneration to diabetes and metastatic cancer. Previous work revealed a crucial role for NUAK1 in supporting viability of tumour cells specifically when MYC is overexpressed. This role is surprising, given that NUAK1 is activated by the tumour suppressor LKB1. Here we show that, in tumour cells lacking LKB1, NUAK1 activity is maintained by an alternative pathway involving calcium-dependent activation of PKCα. Calcium/PKCα-dependent activation of NUAK1 supports engagement of the AMPK-TORC1 metabolic checkpoint, thereby protecting tumour cells from MYC-driven cell death, and indeed, MYC selects for this pathway in part via transcriptional regulation of PKCα and ITPR. Our data point to a novel role for calcium in supporting tumour cell viability and clarify the synthetic lethal interaction between NUAK1 and MYC.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Cálcio/metabolismo , Regulação Neoplásica da Expressão Gênica , Osteossarcoma/patologia , Proteína Quinase C-alfa/metabolismo , Proteínas Quinases/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Apoptose , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Neoplasias Ósseas/genética , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/patologia , Sinalização do Cálcio , Proliferação de Células , Células HeLa , Humanos , Osteossarcoma/genética , Osteossarcoma/metabolismo , Fosforilação , Proteína Quinase C-alfa/genética , Proteínas Quinases/genética , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Repressoras/genética , Células Tumorais Cultivadas
8.
Oncogene ; 37(49): 6368-6382, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30061636

RESUMO

The life expectancy for pancreatic cancer patients has seen no substantial changes in the last 40 years as very few and mostly just palliative treatments are available. As the five years survival rate remains around 5%, the identification of novel pharmacological targets and development of new therapeutic strategies are urgently needed. Here we demonstrate that inhibition of the G protein-coupled receptor GPR55, using genetic and pharmacological approaches, reduces pancreatic cancer cell growth in vitro and in vivo and we propose that this may represent a novel strategy to inhibit pancreatic ductal adenocarcinoma (PDAC) progression. Specifically, we show that genetic ablation of Gpr55 in the KRASWT/G12D/TP53WT/R172H/Pdx1-Cre+/+ (KPC) mouse model of PDAC significantly prolonged survival. Importantly, KPC mice treated with a combination of the GPR55 antagonist Cannabidiol (CBD) and gemcitabine (GEM, one of the most used drugs to treat PDAC), survived nearly three times longer compared to mice treated with vehicle or GEM alone. Mechanistically, knockdown or pharmacologic inhibition of GPR55 reduced anchorage-dependent and independent growth, cell cycle progression, activation of mitogen-activated protein kinase (MAPK) signalling and protein levels of ribonucleotide reductases in PDAC cells. Consistent with this, genetic ablation of Gpr55 reduced proliferation of tumour cells, MAPK signalling and ribonucleotide reductase M1 levels in KPC mice. Combination of CBD and GEM inhibited tumour cell proliferation in KPC mice and it opposed mechanisms involved in development of resistance to GEM in vitro and in vivo. Finally, we demonstrate that the tumour suppressor p53 regulates GPR55 protein expression through modulation of the microRNA miR34b-3p. Our results demonstrate the important role played by GPR55 downstream of p53 in PDAC progression. Moreover our data indicate that combination of CBD and GEM, both currently approved for medical use, might be tested in clinical trials as a novel promising treatment to improve PDAC patients' outcome.


Assuntos
Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/patologia , Receptores de Canabinoides/metabolismo , Animais , Antineoplásicos/farmacologia , Canabidiol/farmacologia , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Camundongos , Camundongos Knockout , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Gencitabina
9.
Nat Commun ; 9(1): 1132, 2018 03 19.
Artigo em Inglês | MEDLINE | ID: mdl-29556067

RESUMO

Many epithelial stem cell populations follow a pattern of stochastic stem cell divisions called 'neutral drift'. It is hypothesised that neutral competition between stem cells protects against the acquisition of deleterious mutations. Here we use a Porcupine inhibitor to reduce Wnt secretion at a dose where intestinal homoeostasis is maintained despite a reduction of Lgr5+ stem cells. Functionally, there is a marked acceleration in monoclonal conversion, so that crypts become rapidly derived from a single stem cell. Stem cells located further from the base are lost and the pool of competing stem cells is reduced. We tested whether this loss of stem cell competition would modify tumorigenesis. Reduction of Wnt ligand secretion accelerates fixation of Apc-deficient cells within the crypt leading to accelerated tumorigenesis. Therefore, ligand-based Wnt signalling influences the number of stem cells, fixation speed of Apc mutations and the speed and likelihood of adenoma formation.


Assuntos
Carcinogênese/metabolismo , Transformação Celular Neoplásica/metabolismo , Mucosa Intestinal/citologia , Mucosa Intestinal/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Via de Sinalização Wnt , Aciltransferases/antagonistas & inibidores , Adenoma/etiologia , Adenoma/metabolismo , Adenoma/patologia , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Carcinogênese/efeitos dos fármacos , Transformação Celular Neoplásica/efeitos dos fármacos , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Inibidores Enzimáticos/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Ligantes , Proteínas de Membrana/antagonistas & inibidores , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Pirazinas/farmacologia , Piridinas/farmacologia , Células-Tronco/efeitos dos fármacos , Via de Sinalização Wnt/efeitos dos fármacos
10.
Mucosal Immunol ; 10(2): 352-360, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27435106

RESUMO

Thiopurines are commonly used drugs in the therapy of Crohn's disease, but unfortunately only show a 30% response rate. The biological basis for the thiopurine response is unclear, thus hampering patient selection prior to treatment. A genetic risk factor associated specifically with Crohn's disease is a variant in ATG16L1 that reduces autophagy. We have previously shown that autophagy is involved in dendritic cell (DC)-T-cell interactions and cytoskeletal regulation. Here we further investigated the role of autophagy in DC cytoskeletal modulation and cellular trafficking. Autophagy-deficient DC displayed loss of filopodia, altered podosome distribution, and increased membrane ruffling, all consistent with increased cellular adhesion. Consequently, autophagy-deficient DC showed reduced migration. The cytoskeletal aberrations were mediated through hyperactivation of Rac1, a known thiopurine target. Indeed thiopurines restored the migratory defects in autophagy-deficient DC. Clinically, the ATG16L1 risk variant associated with increased response to thiopurine treatment in patients with Crohn's disease but not ulcerative colitis. These results suggest that the association between ATG16L1 and Crohn's disease is mediated at least in part through Rac1 hyperactivation and subsequent defective DC migration. As this phenotype can be corrected using thiopurines, ATG16L1 genotyping may be useful in the identification of patients that will benefit most from thiopurine treatment.


Assuntos
Proteínas Relacionadas à Autofagia/metabolismo , Autofagia , Doença de Crohn/imunologia , Células Dendríticas/fisiologia , Proteínas rac1 de Ligação ao GTP/metabolismo , Alelos , Animais , Autofagia/genética , Proteínas Relacionadas à Autofagia/genética , Estruturas da Membrana Celular/patologia , Movimento Celular , Células Cultivadas , Colite Ulcerativa/tratamento farmacológico , Doença de Crohn/tratamento farmacológico , Doença de Crohn/genética , Citoesqueleto/metabolismo , Células Dendríticas/patologia , Feminino , Predisposição Genética para Doença , Humanos , Mercaptopurina/uso terapêutico , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Polimorfismo Genético , RNA Interferente Pequeno/genética , Risco
11.
Cancer Res ; 61(19): 7060-4, 2001 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-11585736

RESUMO

Numerous studies have indicated that exposure to nonsteroidal anti-inflammatory drugs is associated with a lowered risk of colorectal cancer. However, analyses of the effect of aspirin upon tumorigenesis in Apc(Min/+) mice have yielded contrasting results. We show that adult dietary exposure to aspirin does not suppress intestinal tumorigenesis in Apc(Min/+) mice, but that continual exposure from the point of conception does. To test whether this regime could suppress the phenotype of murine models of hereditary nonpolyposis colorectal cancer, Msh2-deficient mice were exposed to aspirin. This did not modify the mutator phenotype of Msh2(-/-) mice, but weakly extended survival. Finally, we analyzed (Apc(Min/+), Msh2(-/-)) mice and found that lifetime aspirin exposure significantly delayed the onset of both intestinal and mammary neoplasia. Thus embryonic and perinatal exposure to aspirin suppresses neoplasia specifically associated with the loss of Apc function, opening a potential window of opportunity for nonsteroidal anti-inflammatory drug intervention.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Proteínas de Ligação a DNA , Genes APC/genética , Neoplasias Intestinais/prevenção & controle , Neoplasias Mamárias Experimentais/prevenção & controle , Proteínas Proto-Oncogênicas/genética , Animais , Pareamento Incorreto de Bases , Neoplasias Colorretais Hereditárias sem Polipose/genética , Neoplasias Colorretais Hereditárias sem Polipose/prevenção & controle , Reparo do DNA , Modelos Animais de Doenças , Feminino , Neoplasias Intestinais/genética , Masculino , Neoplasias Mamárias Experimentais/genética , Camundongos , Camundongos Endogâmicos C57BL , Proteína 2 Homóloga a MutS , Mutação , Gravidez , Efeitos Tardios da Exposição Pré-Natal
12.
Oncogene ; 20(15): 1923-7, 2001 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-11313940

RESUMO

Many reports have shown a link between mismatch repair (MMR) deficiency and loss of normal cell cycle control, particularly loss of G2 arrest. However almost all of these studies utilized transformed cell lines, and thus the involvement of other genes in this phenotype cannot be excluded. We have examined the effects of cisplatin treatment on primary embryo fibroblasts (MEFs) derived from mice in which the MMR gene Msh2 had been inactivated (Msh2(-/-)). This analysis determined that both primary Msh2(-/-) and wild type (WT) fibroblasts exhibited an essentially identical G2 arrest following cisplatin treatment. Similarly, we observed a cisplatin-induced G2 arrest in immortalized MMR deficient (Mlh1(-/-) and Pms2(-/-)) and WT MEFs. p53 deficient primary MEFs (p53(-/-)) exhibited both a clear G2 arrest and an increase in cells with a DNA content of 8N in response to cisplatin. When the Msh2 and p53 defects were combined (p53(-/-)/Msh2(-/-)) the G2 arrest was essentially identical to the p53(-/-) fibroblasts. However, the p53(-/-)/Msh2(-/-) fibroblasts demonstrated a further increase in cells with an 8N DNA content, above that seen in the p53(-/-) fibroblasts. These results suggest that loss of MMR on its own is not enough to overcome G2 arrest following exposure to cisplatin but does play a role in preventing polyploidization, or aberrant DNA reduplication, in the absence of functional p53.


Assuntos
Pareamento Incorreto de Bases , Dano ao DNA , Reparo do DNA , Proteínas de Ligação a DNA , Ploidias , Animais , Linhagem Celular , Cisplatino/farmacologia , Fase G2 , Camundongos , Proteína 2 Homóloga a MutS , Proteínas Proto-Oncogênicas/fisiologia , Proteína Supressora de Tumor p53/fisiologia
13.
Oncogene ; 20(27): 3580-4, 2001 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-11429706

RESUMO

Mice deficient for the mismatch repair (MMR) gene Msh2 show accelerated tumourigenesis and a reduced apoptotic response to DNA damage of methylation type. Here we examine the effect of mutation for Msh2 on in vivo mutation frequencies in the intestine as determined by loss of function at the Dolichos biflorus (Dlb-1) locus. Spontaneous mutation frequencies were scored in cohorts of ageing mice either wild type or mutant for Msh2. In mice less than 1 year old, mutation frequencies were only elevated in Msh2 null mice. However, beyond this age heterozygous Msh2 mice showed significantly higher mutation frequencies than controls. These findings implicate a gene dose dependent requirement for Msh2 in mutation suppression and prompted an analysis of young Msh2 mutants following exposure to DNA damage. Following exposure to N-methyl-N'-nitro-N-nitrosoguanidine (MNNG), Msh2 deficient mice show a reduced apoptotic response and an increase in mutation frequency. Heterozygotes did not differ from controls. Following exposure to cisplatin, no significant elevation was seen in mutation frequencies, even within homozygotes. This is particularly surprising given the association between cisplatin resistance and MMR deficiency. These findings therefore demonstrate a complex reliance upon functional Msh2 in mutation surveillance. We have identified three separate scenarios. First, where retention of both Msh2 alleles over an extended period of time appears critical to the suppression of spontaneous mutation; second, 3 weeks following exposure to MNNG, where only complete loss of Msh2 results in elevated mutation; and finally following cisplatin exposure, where induced levels of mutation are independent of Msh2 status.


Assuntos
Pareamento Incorreto de Bases , Reparo do DNA , Proteínas de Ligação a DNA , Dosagem de Genes , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Cisplatino/toxicidade , Genótipo , Heterozigoto , Metilnitronitrosoguanidina/toxicidade , Camundongos , Camundongos Knockout , Proteína 2 Homóloga a MutS , Mutagênese , Mutação , Proteínas Proto-Oncogênicas/deficiência
14.
Oncogene ; 18(51): 7219-25, 1999 Dec 02.
Artigo em Inglês | MEDLINE | ID: mdl-10602475

RESUMO

We have analysed the pattern of beta-catenin expression by immunohistochemistry in mice singly or multiply mutant for Apc, p53 and Msh2. We observed increased expression of beta-catenin in all intestinal lesions arising on an ApcMin+/- background. In all categories of lesion studied mosaic patterns of beta-catenin expression were observed, with the proportion of cells showing enhanced expression decreasing with increasing lesion size. p53 status did not alter these patterns. We also show that beta-catenin dysregulation marks pancreatic abnormalities occurring in ApcMin+/- and (ApcMin+/-, p53-/-) mice. In these mice both adenomas and adenocarcinomas of the pancreas arose and were characterized by increased expression of beta-catenin. We have extended these analyses to intestinal lesions arising in mice mutant for the mismatch repair gene Msh2. In these mice, increased expression of beta-catenin was again observed. However, in contrast with ApcMin+/- mice, a subset of lesions retained normal expression. Taken together, these findings show that increased expression of beta-catenin is an efficient marker of early neoplastic change in both murine intestine and pancreas in Apc mutant mice. However, we also show that dysregulation of beta-catenin is not an obligate step in the development of intestinal lesions, and therefore that genetic events other than the loss of Apc function may initiate the transition from normal to neoplastic epithelium.


Assuntos
Transformação Celular Neoplásica/genética , Proteínas do Citoesqueleto/genética , Proteínas de Ligação a DNA , Regulação Neoplásica da Expressão Gênica , Genes APC , Intestinos/patologia , Proteínas Proto-Oncogênicas/genética , Transativadores , Proteína da Polipose Adenomatosa do Colo , Animais , Caderinas/genética , Intestinos/fisiologia , Camundongos , Proteína 2 Homóloga a MutS , Mutação , Proteínas Proto-Oncogênicas/deficiência , Proteína Supressora de Tumor p53/genética , beta Catenina
15.
Oncogene ; 34(31): 4118-29, 2015 Jul 30.
Artigo em Inglês | MEDLINE | ID: mdl-25347740

RESUMO

Disruption of Apc (adenomatous polyposis coli) within hepatocytes activates Wnt signalling, perturbs differentiation and ultimately leads to neoplasia. Apc negatively regulates Wnt signalling but is also involved in organizing the cytoskeleton and may have a role in chromosome segregation. In vitro studies have implicated Apc in the control of genomic stability. However, the relevance of this data has been questioned in vivo as Apc is lost earlier than the onset of genomic instability. Here we analyse the relationship between immediate loss of Apc and the acquisition of genomic instability in hepatocytes. We used Cre-lox technology to inactivate Apc and in combination with p53 in vivo, to define the consequences of gene loss on cell cycle regulation, proliferation, death and aneuploidy. We show that, although Apc loss leads to increased proliferation, it also leads to increased apoptosis, the accumulation of p53, p21 and markers of double-strand breaks and DNA repair. Flow cytometry revealed an increased 4N DNA content, consistent with a G2 arrest. Levels of anaphase bridges were also elevated, implicating failed chromosome segregation. This was accompanied by an increase in centrosome number, which demonstrates a role for Apc in maintaining euploidy. To address the role of p53 in these processes, we analysed combined loss of Apc and p53, which led to a further increase in proliferation, cell death, DNA damages and repair and a bypass of G2 arrest than was observed with Apc loss. However, we observed only a marginal effect on anaphase bridges and centrosome number, which could be due to increased cell death. Our data therefore establishes, in an in vivo setting, that APC loss leads to a DNA damage signature and genomic instability in the liver and that additional loss of p53 leads to an increase in the DNA damage signal but not to an immediate increase in the genomic instability phenotype.


Assuntos
Dano ao DNA/genética , Epistasia Genética , Genes APC/fisiologia , Genes p53/fisiologia , Instabilidade Genômica/genética , Hepatócitos/patologia , Animais , Proliferação de Células/genética , Células Cultivadas , Reparo do DNA/genética , Feminino , Hepatomegalia/genética , Hepatomegalia/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , beta-Naftoflavona
17.
Mutat Res ; 452(2): 149-62, 2000 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-11024475

RESUMO

P53 is extremely well characterised as a tumour suppressor gene, and many activities have been attributed to it which are consistent with this function. However, despite being the subject of intense study it still remains unclear precisely which of these functions is crucial to its in vivo role as a tumour suppressor gene. This is particularly true of its role in the induction of apoptosis. The original observation of p53-dependent apoptosis gave rise to the following hypothesis: namely, that p53 deficiency leads to a persistence of DNA damaged cells which are the potential founders of malignancy. This review summarises the data for and against this hypothesis, with specific emphasis on data obtained from studies of the murine intestine. What emerges from these studies is a complex picture, where data can be obtained in support of this hypothesis, but there are many circumstances which exist where it is not supported. Taken together this collection of data suggests that the abrogation of p53-dependent apoptosis may indeed impact upon carcinogenesis and neoplastic progression, but that the simplistic notion of p53 as the single gatekeeper of this pathway is untenable.


Assuntos
Proteínas de Ligação a DNA , Genes p53 , Animais , Apoptose , Sobrevivência Celular , Camundongos , Camundongos Knockout , Proteína 2 Homóloga a MutS , Mutação , Fenótipo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/fisiologia
18.
Oncogene ; 33(25): 3325-33, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-23873029

RESUMO

Many tumours harbour mutations in the p53 tumour-suppressor gene that result in the expression of a mutant p53 protein. This mutant p53 protein has, in most cases, lost wild-type transcriptional activity and can also acquire novel functions in promoting invasion and metastasis. One of the mechanisms underlying these novel functions involves the ability of the mutant p53 to interfere with other transcription factors, including the p53 family protein TAp63. To investigate whether simultaneous depletion of both p53 and TAp63 can recapitulate the effect of mutant p53 expression in vivo, we used a mouse model of pancreatic cancer in which the expression of mutant p53 resulted in the rapid appearance of primary tumours and metastases. As shown previously, loss of one allele of wild-type (WT) p53 accelerated tumour development. A change of one WT p53 allele into mutant p53 did not further accelerate tumour development, but did promote the formation of metastasis. By contrast, loss of TAp63 did not significantly accelerate tumour development or metastasis. However, simultaneous depletion of p53 and TAp63 led to both rapid tumour development and metastatic potential, although the incidence of metastases remained lower than that seen in mutant p53-expressing tumours. TAp63/p53-null cells derived from these mice also showed an enhanced ability to scatter and invade in tissue culture as was observed in mutant p53 cells. These data suggest that depletion of TAp63 in a p53-null tumour can promote metastasis and recapitulate-to some extent-the consequences of mutant p53 expression.


Assuntos
Neoplasias Pancreáticas/patologia , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética , Animais , Linhagem Celular Tumoral , Humanos , Camundongos , Mutação , Metástase Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo
19.
Cell Death Differ ; 21(6): 956-66, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24583641

RESUMO

Recent studies have suggested that C-MYC may be an excellent therapeutic cancer target and a number of new agents targeting C-MYC are in preclinical development. Given most therapeutic regimes would combine C-MYC inhibition with genotoxic damage, it is important to assess the importance of C-MYC function for DNA damage signalling in vivo. In this study, we have conditionally deleted the c-Myc gene in the adult murine intestine and investigated the apoptotic response of intestinal enterocytes to DNA damage. Remarkably, c-Myc deletion completely abrogated the immediate wave of apoptosis following both ionizing irradiation and cisplatin treatment, recapitulating the phenotype of p53 deficiency in the intestine. Consistent with this, c-Myc-deficient intestinal enterocytes did not upregulate p53. Mechanistically, this was linked to an upregulation of the E3 Ubiquitin ligase Mdm2, which targets p53 for degradation in c-Myc-deficient intestinal enterocytes. Further, low level overexpression of c-Myc, which does not impact on basal levels of apoptosis, elicited sustained apoptosis in response to DNA damage, suggesting c-Myc activity acts as a crucial cell survival rheostat following DNA damage. We also identify the importance of MYC during DNA damage-induced apoptosis in several other tissues, including the thymus and spleen, using systemic deletion of c-Myc throughout the adult mouse. Together, we have elucidated for the first time in vivo an essential role for endogenous c-Myc in signalling DNA damage-induced apoptosis through the control of the p53 tumour suppressor protein.


Assuntos
Apoptose/genética , Enterócitos/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/efeitos da radiação , Sobrevivência Celular/efeitos dos fármacos , Cisplatino/administração & dosagem , Dano ao DNA/efeitos dos fármacos , Dano ao DNA/efeitos da radiação , Enterócitos/efeitos dos fármacos , Enterócitos/efeitos da radiação , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , Radiação Ionizante
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA