Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Biosci ; 14(1): 38, 2024 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-38521952

RESUMO

BACKGROUND: Bacterial cancer therapy was first trialled in patients at the end of the nineteenth century. More recently, tumour-targeting bacteria have been harnessed to deliver plasmid-expressed therapeutic interfering RNA to a range of solid tumours. A major limitation to clinical translation of this is the short-term nature of RNA interference in vivo due to plasmid instability. To overcome this, we sought to develop tumour-targeting attenuated bacteria that stably express shRNA by virtue of integration of an expression cassette within the bacterial chromosome and demonstrate therapeutic efficacy in vitro and in vivo. RESULTS: The attenuated tumour targeting Salmonella typhimurium SL7207 strain was modified to carry chromosomally integrated shRNA expression cassettes at the xylA locus. The colorectal cancer cell lines SW480, HCT116 and breast cancer cell line MCF7 were used to demonstrate the ability of these modified strains to perform intracellular infection and deliver effective RNA and protein knockdown of the target gene c-Myc. In vivo therapeutic efficacy was demonstrated using the Lgr5creERT2Apcflx/flx and BlgCreBrca2flx/flp53flx/flx orthotopic immunocompetent mouse models of colorectal and breast cancer, respectively. In vitro co-cultures of breast and colorectal cancer cell lines with modified SL7207 demonstrated a significant 50-95% (P < 0.01) reduction in RNA and protein expression with SL7207/c-Myc targeted strains. In vivo, following establishment of tumour tissue, a single intra-peritoneal administration of 1 × 106 CFU of SL7207/c-Myc was sufficient to permit tumour colonisation and significantly extend survival with no overt toxicity in control animals. CONCLUSIONS: In summary we have demonstrated that tumour tropic bacteria can be modified to safely deliver therapeutic levels of gene knockdown. This technology has the potential to specifically target primary and secondary solid tumours with personalised therapeutic payloads, providing new multi-cancer detection and treatment options with minimal off-target effects. Further understanding of the tropism mechanisms and impact on host immunity and microbiome is required to progress to clinical translation.

2.
Dis Model Mech ; 17(1)2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38149669

RESUMO

LYN kinase is expressed in BRCA1 loss-of-function-dependent mouse mammary tumours, in the cells of origin of such tumours, and in human breast cancer. Suppressing LYN kinase activity in BRCA1-defective cell lines as well as in in vitro cultures of Brca1-null mouse mammary tumours is deleterious to their growth. Here, we examined the interaction between LYN kinase and BRCA1 loss-of-function in an in vivo mouse mammary tumour model, using conditional knockout Brca1 and Lyn alleles. Comparison of Brca1 tumour cohorts showed little difference in mammary tumour formation between animals that were wild type, heterozygous or homozygous for the conditional Lyn allele, although this was confounded by factors including incomplete Lyn recombination in some tumours. RNA-sequencing analysis demonstrated that tumours with high levels of Lyn gene expression had a slower doubling time, but this was not correlated with levels of LYN staining in tumour cells themselves. Rather, high Lyn expression and slower tumour growth were likely a result of B-cell infiltration. The multifaceted role of LYN indicates that it is likely to present difficulties as a therapeutic target in breast cancer.


Assuntos
Neoplasias da Mama , Neoplasias Mamárias Animais , Animais , Feminino , Humanos , Camundongos , Proteína BRCA1/genética , Mama/patologia , Neoplasias da Mama/genética , Linhagem Celular , Neoplasias Mamárias Animais/genética , Neoplasias Mamárias Animais/patologia , Camundongos Knockout
3.
Cancers (Basel) ; 15(17)2023 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-37686600

RESUMO

A better understanding of the mechanisms generating tumour heterogeneity will allow better targeting of current therapies, identify potential resistance mechanisms and highlight new approaches for therapy. We have previously shown that in genetically modified mouse models carrying conditional oncogenic alleles, mammary tumour histotype varies depending on the combination of alleles, the cell type to which they are targeted and, in some cases, reproductive history. This suggests that tumour heterogeneity is not a purely stochastic process; rather, differential activation of signalling pathways leads to reproducible differences in tumour histotype. We propose the NOTCH signalling pathway as one such pathway. Here, we have crossed conditional knockout Notch1 or Notch2 alleles into an established mouse mammary tumour model. Notch1/2 deletion had no effect on tumour-specific survival; however, loss of Notch alleles resulted in a dose-dependent increase in metaplastic adenosquamous carcinomas (ASQCs). ASQCs and adenomyoepitheliomas (AMEs) also demonstrated a significant increase in AKT signalling independent of Notch status. Therefore, the NOTCH pathway is a suppressor of the ASQC phenotype, while increased PI3K/AKT signalling is associated with ASQC and AME tumours. We propose a model in which PI3K/AKT and NOTCH signalling act interact to determine mouse mammary tumour histotype.

4.
BMC Cancer ; 19(1): 677, 2019 Jul 10.
Artigo em Inglês | MEDLINE | ID: mdl-31291912

RESUMO

BACKGROUND: Canonical WNT signalling plays a critical role in the regulation of ovarian development; mis-regulation of this key pathway in the adult ovary is associated with subfertility and tumourigenesis. The roles of Adenomatous polyposis coli 2 (APC2), a little-studied WNT signalling pathway regulator, in ovarian homeostasis, fertility and tumourigenesis have not previously been explored. Here, we demonstrate essential roles of APC2 in regulating ovarian WNT signalling and ovarian homeostasis. METHODS: A detailed analysis of ovarian histology, gene expression, ovulation and hormone levels was carried out in 10 week old and in aged constitutive APC2-knockout (Apc2-/-) mice (mixed background). Statistical significance for qRT-PCR data was determined from 95% confidence intervals. Significance testing was performed using 2-tailed Student's t-test, when 2 experimental cohorts were compared. When more were compared, ANOVA test was used, followed by a post-hoc test (LSD or Games-Howell). P-values of < 0.05 were considered statistically significant. RESULTS: APC2-deficiency resulted in activation of ovarian WNT signalling and sub-fertility driven by intra-ovarian defects. Follicular growth was perturbed, resulting in a reduced rate of ovulation and corpora lutea formation, which could not be rescued by administration of gonadotrophins. Defects in steroidogenesis and follicular vascularity contributed to the subfertility phenotype. Tumour incidence was assessed in aged APC2-deficient mice, which also carried a hypomorphic Apc allele. APC2-deficiency in these mice resulted in predisposition to granulosa cell tumour (GCT) formation, accompanied by acute tumour-associated WNT-signalling activation and a histologic pattern and molecular signature seen in human adult GCTs. CONCLUSIONS: Our work adds APC2 to the growing list of WNT-signalling members that regulate ovarian homeostasis, fertility and suppress GCT formation. Importantly, given that the APC2-deficient mouse develops tumours that recapitulate the molecular signature and histological features of human adult GCTs, this mouse has excellent potential as a pre-clinical model to study ovarian subfertility and transitioning to GCT, tumour biology and for therapeutic testing.


Assuntos
Carcinogênese/metabolismo , Proteínas do Citoesqueleto/metabolismo , Fertilidade , Ovário/metabolismo , Via de Sinalização Wnt , Análise de Variância , Animais , Proteínas do Citoesqueleto/genética , Feminino , Proteína Forkhead Box O1/metabolismo , Técnicas de Inativação de Genes , Tumor de Células da Granulosa/etiologia , Tumor de Células da Granulosa/metabolismo , Homeostase , Infertilidade/etiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Animais , Folículo Ovariano/crescimento & desenvolvimento , Neoplasias Ovarianas/etiologia , Neoplasias Ovarianas/metabolismo , beta Catenina/metabolismo
5.
Oncotarget ; 10(27): 2586-2606, 2019 Apr 05.
Artigo em Inglês | MEDLINE | ID: mdl-31080552

RESUMO

Tumours defective in the DNA homologous recombination repair pathway can be effectively treated with poly (ADP-ribose) polymerase (PARP) inhibitors; these have proven effective in clinical trials in patients with BRCA gene function-defective cancers. However, resistance observed in both pre-clinical and clinical studies is likely to impact on this treatment strategy. Over-expression of phosphoglycoprotein (P-gp) has been previously suggested as a mechanism of resistance to the PARP inhibitor olaparib in mouse models of Brca1/2-mutant breast cancer. Here, we report that in a Brca2 model treated with olaparib, P-gp upregulation is observed but is not sufficient to confer resistance. Furthermore, resistant/relapsed tumours do not show substantial changes in PK/PD of olaparib, do not downregulate PARP1 or re-establish double stranded DNA break repair by homologous recombination, all previously suggested as mechanisms of resistance. However, resistance is strongly associated with epithelial-mesenchymal transition (EMT) and treatment-naïve tumours given a single dose of olaparib upregulate EMT markers within one hour. Therefore, in this model, olaparib resistance is likely a product of an as-yet unidentified mechanism associated with rapid transition to the mesenchymal phenotype.

6.
Oncogene ; 38(16): 3102, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30622341

RESUMO

This article was originally published under standard licence, but has now been made available under a CC BY 4.0 license. The PDF and HTML versions of the paper have been modified accordingly.

7.
Sci Transl Med ; 10(454)2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-30111642

RESUMO

Liver injury results in rapid regeneration through hepatocyte proliferation and hypertrophy. However, after acute severe injury, such as acetaminophen poisoning, effective regeneration may fail. We investigated how senescence may underlie this regenerative failure. In human acute liver disease, and murine models, p21-dependent hepatocellular senescence was proportionate to disease severity and was associated with impaired regeneration. In an acetaminophen injury mouse model, a transcriptional signature associated with the induction of paracrine senescence was observed within 24 hours and was followed by one of impaired proliferation. In mouse genetic models of hepatocyte injury and senescence, we observed transmission of senescence to local uninjured hepatocytes. Spread of senescence depended on macrophage-derived transforming growth factor-ß1 (TGFß1) ligand. In acetaminophen poisoning, inhibition of TGFß receptor 1 (TGFßR1) improved mouse survival. TGFßR1 inhibition reduced senescence and enhanced liver regeneration even when delivered beyond the therapeutic window for treating acetaminophen poisoning. This mechanism, in which injury-induced senescence impairs liver regeneration, is an attractive therapeutic target for developing treatments for acute liver failure.


Assuntos
Senescência Celular , Regeneração Hepática , Fígado/lesões , Fígado/fisiopatologia , Comunicação Parácrina , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Modelos Animais de Doenças , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/patologia , Macrófagos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Necrose , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo
8.
Oncogene ; 37(45): 5913-5925, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29980790

RESUMO

Cancer cells lose homeostatic flexibility because of mutations and dysregulated signaling pathways involved in maintaining homeostasis. Tuberous Sclerosis Complex 1 (TSC1) and TSC2 play a fundamental role in cell homeostasis, where signal transduction through TSC1/TSC2 is often compromised in cancer, leading to aberrant activation of mechanistic target of rapamycin complex 1 (mTORC1). mTORC1 hyperactivation increases the basal level of endoplasmic reticulum (ER) stress via an accumulation of unfolded protein, due to heightened de novo protein translation and repression of autophagy. We exploit this intrinsic vulnerability of tumor cells lacking TSC2, by treating with nelvinavir to further enhance ER stress while inhibiting the proteasome with bortezomib to prevent effective protein removal. We show that TSC2-deficient cells are highly dependent on the proteosomal degradation pathway for survival. Combined treatment with nelfinavir and bortezomib at clinically relevant drug concentrations show synergy in selectively killing TSC2-deficient cells with limited toxicity in control cells. This drug combination inhibited tumor formation in xenograft mouse models and patient-derived cell models of TSC and caused tumor spheroid death in 3D culture. Importantly, 3D culture assays differentiated between the cytostatic effects of the mTORC1 inhibitor, rapamycin, and the cytotoxic effects of the nelfinavir/bortezomib combination. Through RNA sequencing, we determined that nelfinavir and bortezomib tip the balance of ER protein homeostasis of the already ER-stressed TSC2-deficient cells in favor of cell death. These findings have clinical relevance in stratified medicine to treat tumors that have compromised signaling through TSC and are inflexible in their capacity to restore ER homeostasis.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Neoplasias/patologia , Proteína 2 do Complexo Esclerose Tuberosa/metabolismo , Animais , Bortezomib/farmacologia , Linhagem Celular Tumoral , Estresse do Retículo Endoplasmático/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Nelfinavir/farmacologia , Neoplasias/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Nat Cell Biol ; 17(8): 971-983, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26192438

RESUMO

Hepatocytes and cholangiocytes self-renew following liver injury. Following severe injury hepatocytes are increasingly senescent, but whether hepatic progenitor cells (HPCs) then contribute to liver regeneration is unclear. Here, we describe a mouse model where the E3 ubiquitin ligase Mdm2 is inducibly deleted in more than 98% of hepatocytes, causing apoptosis, necrosis and senescence with nearly all hepatocytes expressing p21. This results in florid HPC activation, which is necessary for survival, followed by complete, functional liver reconstitution. HPCs isolated from genetically normal mice, using cell surface markers, were highly expandable and phenotypically stable in vitro. These HPCs were transplanted into adult mouse livers where hepatocyte Mdm2 was repeatedly deleted, creating a non-competitive repopulation assay. Transplanted HPCs contributed significantly to restoration of liver parenchyma, regenerating hepatocytes and biliary epithelia, highlighting their in vivo lineage potency. HPCs are therefore a potential future alternative to hepatocyte or liver transplantation for liver disease.


Assuntos
Ductos Biliares/transplante , Linhagem da Célula , Proliferação de Células , Células Epiteliais/transplante , Hepatócitos/transplante , Regeneração Hepática , Fígado , Transplante de Células-Tronco , Células-Tronco , Animais , Apoptose , Ductos Biliares/metabolismo , Ductos Biliares/patologia , Biomarcadores/metabolismo , Separação Celular , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Genótipo , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/metabolismo , Fígado/patologia , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Necrose , Fenótipo , Proteínas Proto-Oncogênicas c-mdm2/deficiência , Proteínas Proto-Oncogênicas c-mdm2/genética , Células-Tronco/metabolismo , Células-Tronco/patologia , Fatores de Tempo
10.
Mol Cancer Ther ; 14(5): 1259-69, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25695957

RESUMO

In this article, we report the development and preclinical validation of combinatorial therapy for treatment of cancers using RNA interference (RNAi). RNAi technology is an attractive approach to silence genes responsible for disease onset and progression. Currently, the critical challenge facing the clinical success of RNAi technology is in the difficulty of delivery of RNAi inducers, due to low transfection efficiency, difficulties of integration into host DNA and unstable expression. Using the macromolecule polyglycidal methacrylate (PGMA) as a platform to graft multiple polyethyleneimine (PEI) chains, we demonstrate effective delivery of small oligos (anti-miRs and mimics) and larger DNAs (encoding shRNAs) in a wide variety of cancer cell lines by successful silencing/activation of their respective target genes. Furthermore, the effectiveness of this therapy was validated for in vivo tumor suppression using two transgenic mouse models; first, tumor growth arrest and increased animal survival was seen in mice bearing Brca2/p53-mutant mammary tumors following daily intratumoral treatment with nanoparticles conjugated to c-Myc shRNA. Second, oral delivery of the conjugate to an Apc-deficient crypt progenitor colon cancer model increased animal survival and returned intestinal tissue to a non-wnt-deregulated state. This study demonstrates, through careful design of nonviral nanoparticles and appropriate selection of therapeutic gene targets, that RNAi technology can be made an affordable and amenable therapy for cancer.


Assuntos
Neoplasias da Mama/terapia , Neoplasias Colorretais/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Terapêutica com RNAi/métodos , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Feminino , Inativação Gênica , Células HEK293 , Humanos , Células Jurkat , Células MCF-7 , Camundongos , Células NIH 3T3 , Nanoconjugados , Transplante de Neoplasias , Polipropilenos/química , Interferência de RNA
11.
Cancer Res ; 69(9): 3850-5, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19383921

RESUMO

Germ-line heterozygosity of the BRCA2 gene in women predisposes to breast and ovarian cancers. Successful therapies targeted specifically at these neoplasms have thus far remained elusive. Recent studies in mice have shown that inhibition of poly(ADP-ribose) polymerase-1 (PARP-1) targets cells lacking Brca2 and xenografts derived from BRCA2-deficient ES cells or Chinese hamster ovary cells. We set out to develop a more relevant preclinical model that will inform and accelerate translation into the clinic. As such, we conditionally deleted Brca2 and p53 within murine mammary epithelium and treated the resulting tumors in situ with a highly potent PARP-1 inhibitor (AZD2281) alone or in combination with carboplatin. Daily exposure to AZD2281 for 28 days caused significant regression or growth inhibition in 46 of 52 tumors. This response was shown to be specific to tumors lacking both Brca2and p53. AZD2281/carboplatin combination therapy for 28 days showed no advantage over carboplatin monotherapy. However, if PARP inhibitor treatment was continued, this significantly increased the time to tumor relapse and death in these mice. This preclinical study is the first to show in vivo hypersensitivity of spontaneously arising Brca2-deficient mammary tumors to PARP-1 inhibition monotherapy or combination therapy. As such, our data add substantial weight to the argument for the use of PARP inhibitors as therapeutic agents against human breast cancers in which BRCA2 is mutated. Moreover, the specificity that we have shown further suggests that PARP inhibitors will be generally effective against tumors caused by dysregulation of components of the homologous recombination pathway.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Genes BRCA2 , Genes p53 , Neoplasias Mamárias Experimentais/tratamento farmacológico , Neoplasias Mamárias Experimentais/genética , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Proteína BRCA2/deficiência , Proteína BRCA2/genética , Carboplatina/administração & dosagem , Modelos Animais de Doenças , Feminino , Neoplasias Mamárias Experimentais/enzimologia , Camundongos , Camundongos Transgênicos , Mutação , Ftalazinas/administração & dosagem , Piperazinas/administração & dosagem , Poli(ADP-Ribose) Polimerase-1 , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
12.
Cancer Res ; 65(22): 10145-8, 2005 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-16287996

RESUMO

The genes encoding the BRCA1 and BRCA2 tumor suppressors are the most commonly mutated in human familial breast cancers. Both have separate roles in the maintenance of genomic stability through involvement in homologous recombination, an error-free process enabling cells to repair DNA double-strand breaks. We have previously shown that cre-mediated conditional deletion of Brca2 within the mouse small intestine sensitizes the tissue to DNA damage. Eventually, the tissue repopulates via stem cells in which recombination at the floxed Brca2 allele has not taken place. In this study, we have treated Brca2-deficient small intestine with a potent small-molecule inhibitor of poly(ADP-ribose) polymerase 1 (PARP1), an enzyme predominantly involved in the recognition of DNA single-strand breaks. Brca2 deficiency rendered otherwise normal cells exquisitely sensitive to PARP inhibition, resulting in very high levels of apoptosis as early as 6 hours after treatment, with evidence for repopulation of the tissue at 12 hours. Furthermore, the intestines of animals treated with serial injections of the inhibitor repopulated very rapidly in comparison with those from untreated mice. Our results represent the first in vivo demonstration that inhibition of PARP1 activity confers exquisite sensitivity to death in physiologically normal Brca2-deficient cells, suggesting that such a regimen may be extremely potent prophylactically in women heterozygous for the BRCA2 gene, as well as against established tumors lacking functional BRCA2.


Assuntos
Proteína BRCA2/deficiência , Neoplasias da Mama/prevenção & controle , Deleção de Genes , Genes BRCA2 , Mucosa Intestinal/fisiologia , Intestino Delgado/fisiologia , Inibidores de Poli(ADP-Ribose) Polimerases , Animais , Apoptose/efeitos dos fármacos , Disponibilidade Biológica , Neoplasias da Mama/genética , Dano ao DNA , Inibidores Enzimáticos/farmacocinética , Inibidores Enzimáticos/farmacologia , Fluorbenzenos/farmacocinética , Fluorbenzenos/farmacologia , Mucosa Intestinal/efeitos dos fármacos , Mucosa Intestinal/enzimologia , Mucosa Intestinal/metabolismo , Intestino Delgado/efeitos dos fármacos , Intestino Delgado/enzimologia , Intestino Delgado/metabolismo , Camundongos , Camundongos Transgênicos , Ftalazinas/farmacocinética , Ftalazinas/farmacologia
13.
Oncogene ; 24(23): 3842-6, 2005 May 26.
Artigo em Inglês | MEDLINE | ID: mdl-15735671

RESUMO

The gene encoding the human BRCA2 tumour suppressor is mutated in a number of different tumour types, most notably inherited breast cancers. The primary role of BRCA2 is thought to lie in the maintenance of genomic stability via its role in the homologous recombination pathway. We generated mice in which Brca2 was deleted from virtually all cells within the adult small intestine, using a CYP1A1-driven Cre-Lox approach. We noted a significant p53-dependent increase in the levels of spontaneous apoptosis which persisted for several months after removal of the gene and ultimately we observed the spontaneous deletion of Brca2-deficient stem cells. Brca2 deficiency did not lead to gross changes in intestinal physiology but did enhance sensitivity to a variety of DNA crosslinking agents. Taken together, our results indicate that Brca2 plays an important role in the response to DNA damage in the small intestine. Furthermore, we show that Brca2 deficiency results in the spontaneous deletion of stem cells, thereby protecting the small intestine against tumorigenesis.


Assuntos
Apoptose , Proteína BRCA2/fisiologia , Intestino Delgado/patologia , Células-Tronco/patologia , Proteína Supressora de Tumor p53/fisiologia , Animais , Proteína BRCA2/deficiência , Dano ao DNA , Camundongos
14.
Cancer Res ; 65(2): 410-6, 2005 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-15695381

RESUMO

Mutations of Apc and p53 have both been implicated in human and murine mammary neoplasia. To investigate potential interactions between Apc and p53, we conditionally inactivated Apc in both the presence and the absence of functional p53. Apc deficiency on its own leads to the development of metaplasia but not neoplasia. We show here that these areas of metaplasia are characterized by elevated levels of both p53 and p21. In the additional absence of p53,there is rapid progression to neoplasia, with 44.4% of lymphoma-free mice developing a mammary tumor with earliest observed onset at pregnancy. To investigate the mechanism by which p53 deficiency accelerates neoplasia, we used the Rosa26R reporter strain as a marker of Cre-mediated recombination and show a role for p53 in the loss of Apc-deficient cells. This role seems limited to pregnancy and subsequent time points. We therefore show clear synergy between these two mutations in mammary gland neoplasia and present data to suggest that at least one mechanism for this acceleration is the p53-dependent loss of Apc-deficient cells.


Assuntos
Transformação Celular Neoplásica/genética , Genes APC , Genes p53/genética , Neoplasias Mamárias Experimentais/genética , Proteína da Polipose Adenomatosa do Colo/biossíntese , Proteína da Polipose Adenomatosa do Colo/deficiência , Proteína da Polipose Adenomatosa do Colo/genética , Animais , Proteínas de Ciclo Celular/biossíntese , Proteínas de Ciclo Celular/genética , Inibidor de Quinase Dependente de Ciclina p21 , Proteínas do Citoesqueleto/biossíntese , Proteínas do Citoesqueleto/genética , Feminino , Mutação da Fase de Leitura , Inativação Gênica , Glândulas Mamárias Animais/patologia , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Metaplasia/genética , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Gravidez , Transativadores/biossíntese , Transativadores/genética , Proteína Supressora de Tumor p53/biossíntese , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética , Regulação para Cima , beta Catenina
15.
Oncogene ; 21(42): 6446-57, 2002 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-12226748

RESUMO

Apc (adenomatous polyposis coli) encodes a tumour suppressor gene that is mutated in the majority of colorectal cancers. Recent evidence has also implicated Apc mutations in the aetiology of breast tumours. Apc is a component of the canonical Wnt signal transduction pathway, of which one target is Tcf-1. In the mouse, mutations of both Apc and Tcf-1 have been implicated in mammary tumorigenesis. We have conditionally inactivated Apc in both the presence and absence of Tcf-1 to examine the function of these genes in both normal and neoplastic development. Mice harbouring mammary-specific mutations in Apc show markedly delayed development of the mammary ductal network. During lactation, the mice develop multiple metaplastic growths which, surprisingly, do not spontaneously progress to neoplasia up to a year following their induction. However, additional deficiency of Tcf-1 completely blocks normal mammary development and results in acanthoma.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Mama/crescimento & desenvolvimento , Proteínas de Ligação a DNA/deficiência , Neoplasias Mamárias Experimentais/genética , Metaplasia/patologia , Neoplasias Cutâneas/patologia , Fatores de Transcrição/deficiência , Animais , Carcinoma de Células Acinares/patologia , Carcinoma de Células Escamosas/patologia , Ciclina D1/metabolismo , Proteínas do Citoesqueleto/metabolismo , Modelos Animais de Doenças , Feminino , Inativação Gênica , Genes myc/fisiologia , Genótipo , Mutação em Linhagem Germinativa , Fator 1-alfa Nuclear de Hepatócito , Técnicas Imunoenzimáticas , Integrases , Óperon Lac/fisiologia , Fator 1 de Ligação ao Facilitador Linfoide , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos , Fenótipo , Fator 1 de Transcrição de Linfócitos T , Transativadores/metabolismo , Proteínas Virais , beta Catenina
16.
Mol Cell Biol ; 22(17): 6170-82, 2002 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-12167711

RESUMO

The Mdm2 protein mediates ubiquitylation and degradation of p53 and is a key regulator of this tumor suppressor. More recently, it has been shown that Mdm2 is highly phosphorylated within its central acidic domain. In order to address the issue of how these modifications might regulate Mdm2 function, putative phosphorylation sites within this domain were substituted, individually or in pairs, with alanine residues. Mutants with serine-to-alanine substitutions between residues 244 and 260 abolished or at least reduced the capacity of Mdm2 to promote p53 degradation. In each case, loss of degradation function was independent of the ability to bind to p53 or p14ARF. Moreover, each of the Mdm2 mutants completely retained the capacity to act as a ubiquitin ligase in vivo. Thus, ubiquitylation and degradation can be uncoupled. Two-dimensional phosphopeptide mapping coupled with the use of phospho-specific antibodies revealed that Mdm2 is phosphorylated physiologically at several sites within this region, consistent with the idea that phosphorylation is important for Mdm2 activity. Strikingly, treatment of cells with ionizing radiation resulted in a significant decrease in the phosphorylation of residues that are important for p53 turnover. This hypophosphorylation preceded p53 accumulation. These findings indicate that Mdm2 contributes an additional function toward the degradation of p53 that is distinct from its ubiquitin ligase activity and is regulated by phosphorylation. Our model suggests that hypophosphorylation of Mdm2 in response to ionizing irradiation inactivates this novel function, thereby contributing to p53 stabilization.


Assuntos
Proteínas Nucleares , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas/fisiologia , Proteína Supressora de Tumor p53/química , Sequência de Aminoácidos , Animais , Sítios de Ligação , Células COS , Carcinoma/patologia , Células Cultivadas/metabolismo , Células Cultivadas/efeitos da radiação , Chlorocebus aethiops , Cisteína Endopeptidases/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efeitos da radiação , Raios gama , Humanos , Neoplasias Pulmonares/patologia , Camundongos , Dados de Sequência Molecular , Complexos Multienzimáticos/metabolismo , Mutagênese Sítio-Dirigida , Mapeamento de Peptídeos , Fosforilação , Complexo de Endopeptidases do Proteassoma , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas/química , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas c-mdm2 , Proteínas Recombinantes de Fusão/fisiologia , Deleção de Sequência , Especificidade da Espécie , Transfecção , Células Tumorais Cultivadas , Proteína Supressora de Tumor p14ARF/metabolismo , Ubiquitina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...