Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 2665, 2022 05 13.
Artigo em Inglês | MEDLINE | ID: mdl-35562376

RESUMO

Pancreatic intraepithelial neoplasia (PanIN) is a precursor of pancreatic ductal adenocarcinoma (PDAC), which commonly occurs in the general populations with aging. Although most PanIN lesions (PanINs) harbor oncogenic KRAS mutations that initiate pancreatic tumorigenesis; PanINs rarely progress to PDAC. Critical factors that promote this progression, especially targetable ones, remain poorly defined. We show that peroxisome proliferator-activated receptor-delta (PPARδ), a lipid nuclear receptor, is upregulated in PanINs in humans and mice. Furthermore, PPARδ ligand activation by a high-fat diet or GW501516 (a highly selective, synthetic PPARδ ligand) in mutant KRASG12D (KRASmu) pancreatic epithelial cells strongly accelerates PanIN progression to PDAC. This PPARδ activation induces KRASmu pancreatic epithelial cells to secrete CCL2, which recruits immunosuppressive macrophages and myeloid-derived suppressor cells into pancreas via the CCL2/CCR2 axis to orchestrate an immunosuppressive tumor microenvironment and subsequently drive PanIN progression to PDAC. Our data identify PPARδ signaling as a potential molecular target to prevent PDAC development in subjects harboring PanINs.


Assuntos
Carcinoma in Situ , Carcinoma Ductal Pancreático , PPAR delta , Neoplasias Pancreáticas , Animais , Carcinogênese/genética , Carcinogênese/patologia , Carcinoma in Situ/patologia , Carcinoma Ductal Pancreático/patologia , Humanos , Ligantes , Camundongos , PPAR delta/genética , Pâncreas/patologia , Neoplasias Pancreáticas/patologia , Proteínas Proto-Oncogênicas p21(ras)/genética , Microambiente Tumoral/genética , Neoplasias Pancreáticas
2.
SLAS Discov ; 27(3): 175-184, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35314378

RESUMO

High-throughput viability screens are commonly used in the identification and development of chemotherapeutic drugs. These systems rely on the fidelity of the cellular model systems to recapitulate the drug response that occurs in vivo. In recent years, there has been an expansion in the utilization of patient-derived materials as well as advanced cell culture techniques, such as multi-cellular tumor organoids, to further enhance the translational relevance of cellular model systems. Simple quantitative analysis remains a challenge, primarily due to the difficulties of robust image segmentation in heterogenous 3D cultures. However, explicit segmentation is not required with the advancement of deep learning, and it can be used for both continuous (regression) or categorical classification problems. Deep learning approaches are additionally benefited by being fully data-driven and highly automatable, thus they can be established and run with minimal to no user-defined parameters. In this article, we describe the development and implementation of a regressive deep learning model trained on brightfield images of patient-derived organoids and use the terminal viability readout (CellTiter-Glo) as training labels. Ultimately, this has led to the generation of a non-invasive and label-free tool to evaluate changes in organoid viability.


Assuntos
Técnicas de Cultura de Células , Organoides , Sobrevivência Celular , Humanos
3.
Cell Rep ; 32(7): 108049, 2020 08 18.
Artigo em Inglês | MEDLINE | ID: mdl-32814052

RESUMO

APC mutation activation of Wnt/ß-catenin drives initiation of colorectal carcinogenesis (CRC). Additional factors potentiate ß-catenin activation to promote CRC. Western diets are enriched in linoleic acid (LA); LA-enriched diets promote chemically induced CRC in rodents. 15-Lipoxygenase-1 (15-LOX-1), the main LA-metabolizing enzyme, is transcriptionally silenced during CRC. Whether LA and 15-LOX-1 affect Wnt/ß-catenin signaling is unclear. We report that high dietary LA promotes CRC in mice treated with azoxymethane or with an intestinally targeted Apc mutation (ApcΔ580) by upregulating Wnt receptor LRP5 protein expression and ß-catenin activation. 15-LOX-1 transgenic expression in mouse intestinal epithelial cells suppresses LRP5 protein expression, ß-catenin activation, and CRC. 15-LOX-1 peroxidation of LA in phosphatidylinositol-3-phosphates (PI3P_LA) leads to PI3P_13-HODE formation, which decreases PI3P binding to SNX17 and LRP5 and inhibits LRP5 recycling from endosomes to the plasma membrane, thereby increasing LRP5 lysosomal degradation. This regulatory mechanism of LRP5/Wnt/ß-catenin signaling could be therapeutically targeted to suppress CRC.


Assuntos
Neoplasias do Colo/genética , Ácido Linoleico/metabolismo , Proteína-5 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Via de Sinalização Wnt/genética , beta Catenina/metabolismo , Animais , Humanos , Camundongos , Transdução de Sinais , Transfecção
4.
Gastroenterology ; 157(1): 163-178, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30885780

RESUMO

BACKGROUND & AIMS: The peroxisome proliferator-activated receptor delta (PPARD) regulates cell metabolism, proliferation, and inflammation and has been associated with gastric and other cancers. Villin-positive epithelial cells are a small population of quiescent gastric progenitor cells. We expressed PPARD from a villin promoter to investigate the role of these cells and PPARD in development of gastric cancer. METHODS: We analyzed gastric tissues from mice that express the Ppard (PPARD1 and PPARD2 mice) from a villin promoter, and mice that did not carry this transgene (controls), by histology and immunohistochemistry. We performed cell lineage-tracing experiments and analyzed the microbiomes, chemokine and cytokine production, and immune cells and transcriptomes of stomachs of these mice. We also performed immunohistochemical analysis of PPARD levels in 2 sets of human gastric tissue microarrays. RESULTS: Thirty-eight percent of PPARD mice developed spontaneous, invasive gastric adenocarcinomas, with severe chronic inflammation. Levels of PPARD were increased in human gastric cancer tissues, compared with nontumor tissues, and associated with gastric cancer stage and grade. We found an inverse correlation between level of PPARD in tumor tissue and patient survival time. Gastric microbiomes from PPARD and control mice did not differ significantly. Lineage-tracing experiments identified villin-expressing gastric progenitor cells (VGPCs) as the origin of gastric tumors in PPARD mice. In these mice, PPARD up-regulated CCL20 and CXCL1, which increased infiltration of the gastric mucosa by immune cells. Immune cell production of inflammatory cytokines promoted chronic gastric inflammation and expansion and transformation of VGPCs, leading to tumorigenesis. We identified a positive-feedback loop between PPARD and interferon gamma signaling that sustained gastric inflammation to induce VGPC transformation and gastric carcinogenesis. CONCLUSIONS: We found PPARD overexpression in VPGCs to result in inflammation, dysplasia, and tumor formation. PPARD and VGPCs might be therapeutic targets for stomach cancer.


Assuntos
Carcinogênese/genética , Transformação Celular Neoplásica/genética , Citocinas/imunologia , Mucosa Gástrica/metabolismo , Interferon gama/imunologia , Receptores Citoplasmáticos e Nucleares/genética , Células-Tronco/metabolismo , Estômago/imunologia , Adenocarcinoma/genética , Adenocarcinoma/imunologia , Animais , Carcinogênese/imunologia , Linhagem da Célula , Transformação Celular Neoplásica/imunologia , Quimiocina CCL20/metabolismo , Quimiocina CXCL1/metabolismo , Quimiocinas , Retroalimentação Fisiológica , Perfilação da Expressão Gênica , Inflamação , Camundongos , Microbiota/imunologia , Proteínas dos Microfilamentos/genética , Células-Tronco/imunologia , Estômago/microbiologia , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia
5.
Cancer Res ; 79(5): 954-969, 2019 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-30679176

RESUMO

APC mutations activate aberrant ß-catenin signaling to drive initiation of colorectal cancer; however, colorectal cancer progression requires additional molecular mechanisms. PPAR-delta (PPARD), a downstream target of ß-catenin, is upregulated in colorectal cancer. However, promotion of intestinal tumorigenesis following deletion of PPARD in Apcmin mice has raised questions about the effects of PPARD on aberrant ß-catenin activation and colorectal cancer. In this study, we used mouse models of PPARD overexpression or deletion combined with APC mutation (ApcΔ580 ) in intestinal epithelial cells (IEC) to elucidate the contributions of PPARD in colorectal cancer. Overexpression or deletion of PPARD in IEC augmented or suppressed ß-catenin activation via up- or downregulation of BMP7/TAK1 signaling and strongly promoted or suppressed colorectal cancer, respectively. Depletion of PPARD in human colorectal cancer organoid cells inhibited BMP7/ß-catenin signaling and suppressed organoid self-renewal. Treatment with PPARD agonist GW501516 enhanced colorectal cancer tumorigenesis in ApcΔ580 mice, whereas treatment with PPARD antagonist GSK3787 suppressed tumorigenesis. PPARD expression was significantly higher in human colorectal cancer-invasive fronts versus their paired tumor centers and adenomas. Reverse-phase protein microarray and validation studies identified PPARD-mediated upregulation of other proinvasive pathways: connexin 43, PDGFRß, AKT1, EIF4G1, and CDK1. Our data demonstrate that PPARD strongly potentiates multiple tumorigenic pathways to promote colorectal cancer progression and invasiveness. SIGNIFICANCE: These findings address long-standing, important, and unresolved questions related to the potential role of PPARD in APC mutation-dependent colorectal tumorigenesis by showing PPARD activation enhances APC mutation-dependent tumorigenesis.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , PPAR delta/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Benzamidas/farmacologia , Carcinogênese , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Progressão da Doença , Células HCT116 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Invasividade Neoplásica , PPAR delta/biossíntese , PPAR delta/genética , Receptores Citoplasmáticos e Nucleares/biossíntese , Receptores Citoplasmáticos e Nucleares/genética , Sulfonas/farmacologia , Tiazóis/farmacologia
6.
JCI Insight ; 2(1): e91419, 2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28097239

RESUMO

Peroxisome proliferator-activated receptor-δ (PPARD) is upregulated in many major human cancers, but the role that its expression in cancer cells has in metastasis remains poorly understood. Here, we show that specific PPARD downregulation or genetic deletion of PPARD in cancer cells significantly repressed metastasis in various cancer models in vivo. Mechanistically, PPARD promoted angiogenesis via interleukin 8 in vivo and in vitro. Analysis of transcriptome profiling of HCT116 colon cancer cells with or without genetic deletion of PPARD and gene expression patterns in The Cancer Genome Atlas colorectal adenocarcinoma database identified novel pro-metastatic genes (GJA1, VIM, SPARC, STC1, SNCG) as PPARD targets. PPARD expression in cancer cells drastically affected epithelial-mesenchymal transition, migration, and invasion, further underscoring its necessity for metastasis. Clinically, high PPARD expression in various major human cancers (e.g., colorectal, lung, breast) was associated with significantly reduced metastasis-free survival. Our results demonstrate that PPARD, a druggable protein, is an important molecular target in metastatic cancer.


Assuntos
Perfilação da Expressão Gênica/métodos , Neoplasias Pulmonares/genética , Metástase Neoplásica/genética , Neoplasias/genética , PPAR delta/genética , Indutores da Angiogênese/metabolismo , Animais , Regulação para Baixo/genética , Transição Epitelial-Mesenquimal/genética , Feminino , Deleção de Genes , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HCT116/citologia , Células HCT116/metabolismo , Humanos , Interleucina-8/metabolismo , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/secundário , Camundongos , Terapia de Alvo Molecular/métodos , Metástase Neoplásica/tratamento farmacológico , Neoplasias/metabolismo , Neoplasias/patologia , PPAR delta/metabolismo
7.
FASEB J ; 29(6): 2359-70, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25713055

RESUMO

The IL-6/signal transducer and activator of transcription 3 (STAT3) pathway is a critical signaling pathway for colitis-associated colorectal cancer (CAC). Peroxisome proliferator-activated receptor (PPAR)-δ, a lipid nuclear receptor, up-regulates IL-6. 15-Lipoxygenase-1 (15-LOX-1), which is crucial to production of lipid signaling mediators to terminate inflammation, down-regulates PPAR-δ. 15-LOX-1 effects on IL-6/STAT3 signaling and CAC tumorigenesis have not been determined. We report that intestinally targeted transgenic 15-LOX-1 expression in mice inhibited azoxymethane- and dextran sodium sulfate-induced CAC, IL-6 expression, STAT3 phosphorylation, and IL-6/STAT3 downstream target (Notch3 and MUC1) expression. 15-LOX-1 down-regulation was associated with IL-6 up-regulation in human colon cancer mucosa. Reexpression of 15-LOX-1 in human colon cancer cells suppressed IL-6 mRNA expression, STAT3 phosphorylation, IL-6 promoter activity, and PPAR-δ mRNA and protein expression. PPAR-δ overexpression in colonic epithelial cells promoted CAC tumorigenesis in mice and increased IL-6 expression and STAT3 phosphorylation, whereas concomitant 15-LOX-1 expression in colonic epithelial cells (15-LOX-1-PPAR-δ-Gut mice) suppressed these effects: the number of tumors per mouse (mean ± sem) was 4.22 ± 0.68 in wild-type littermates, 6.67 ± 0.83 in PPAR-δ-Gut mice (P = 0.026), and 2.25 ± 0.25 in 15-LOX-1-PPAR-δ-Gut mice (P = 0.0006). Identification of 15-LOX-1 suppression of PPAR-δ to inhibit IL-6/STAT3 signaling-driven CAC tumorigenesis provides mechanistic insights that can be used to molecularly target CAC.


Assuntos
Araquidonato 15-Lipoxigenase/metabolismo , Colite/metabolismo , Neoplasias do Colo/metabolismo , Interleucina-6/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Araquidonato 15-Lipoxigenase/genética , Azoximetano , Western Blotting , Células CACO-2 , Linhagem Celular Tumoral , Proliferação de Células/genética , Colite/induzido quimicamente , Colite/genética , Neoplasias do Colo/induzido quimicamente , Neoplasias do Colo/genética , Sulfato de Dextrana , Expressão Gênica , Células HCT116 , Humanos , Imuno-Histoquímica , Interleucina-6/genética , Camundongos Transgênicos , PPAR delta/genética , PPAR delta/metabolismo , Fosforilação , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/genética
8.
J Natl Cancer Inst ; 106(4): dju052, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24681603

RESUMO

BACKGROUND: The nuclear receptor peroxisome proliferator-activated receptor-δ/ß (PPAR-d) is upregulated in human colorectal cancers, but its role in colonic tumorigenesis remains controversial. METHODS: We generated a novel mouse model of intestinally targeted PPAR-d overexpression to simulate PPAR-d upregulation in human colon carcinogenesis. Colon-specific PPAR-d overexpression was confirmed by real-time reverse transcription polymerase chain reaction, immunoblotting, and activity assays. Mice with and without targeted PPAR-d overexpression were tested for azoxymethane (AOM)-induced colonic tumorigenesis. Mouse whole-genome transcriptome microarray analyses were performed to identify PPAR-d target genes to promote tumorigenesis. We used linear models to test for PPAR-d overexpression trend effects on tumor multiplicity. All statistical tests were two-sided. RESULTS: Targeted PPAR-d overexpression markedly increased colonic tumor incidence (from 0 of 10 wild-type [WT] littermate mice to 9 of 10 mice [P < .001] in 2 FVB/N background mouse lines [villin-PPAR-d-1 and villin-PPAR-d-2] at a 5-mg/kg AOM dose) and multiplicity (number of tumors per mouse per mg/kg dose of AOM increased from 0.47 [95% confidence interval [CI] = 0.22 to 0.72] for the WT littermates to 2.15 [95% CI = 1.90 to 2.40] [P < .001] for the villin-PPAR-d-1 mice and from 0.44 [95% CI = 0.09 to 0.79] for the WT littermates to 1.91 [95% CI = 1.57 to 2.25] [P < .001] for the villin-PPAR-d-2 mice). PPAR-d overexpression reversed resistance to AOM-induced colonic tumorigenesis in C57BL/6 mice. PPAR-d overexpression modulated expression of several novel PPAR-d target genes in normal-appearing colonic epithelial cells of mice with PPAR-d overexpression in a pattern that matched the changes in colonic tumors. CONCLUSIONS: Our finding that PPAR-d upregulation profoundly enhances susceptibility to colonic tumorigenesis should impact the development of strategies of molecularly targeting PPAR-d in cancer and noncancerous diseases.


Assuntos
Neoplasias do Colo/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Animais , Azoximetano , Carcinógenos , Neoplasias do Colo/induzido quimicamente , Suscetibilidade a Doenças , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Experimentais/metabolismo , PPAR beta/metabolismo , Receptores Citoplasmáticos e Nucleares/genética , Regulação para Cima
9.
Cancer Med ; 3(3): 472-84, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24634093

RESUMO

The expression of 15-lipoxygenase-1 (15-LOX-1) is downregulated in colon cancer and other major cancers, and 15-LOX-1 reexpression in cancer cells suppresses colonic tumorigenesis. Various lines of evidence indicate that 15-LOX-1 expression suppresses premetastatic stages of colonic tumorigenesis; nevertheless, the role of 15-LOX-1 loss of expression in cancer epithelial cells in metastases continues to be debated. Hypoxia, a common feature of the cancer microenvironment, promotes prometastatic mechanisms such as the upregulation of hypoxia-inducible factor (HIF)-1α, a transcriptional master regulator that enhances cancer cell metastatic potential, angiogenesis, and tumor cell invasion and migration. We have, therefore, tested whether restoring 15-LOX-1 in colon cancer cells affects cancer cells' hypoxia response that promotes metastasis. We found that 15-LOX-1 reexpression in HCT116, HT29LMM, and LoVo colon cancer cells inhibited survival, vascular endothelial growth factor (VEGF) expression, angiogenesis, cancer cell migration and invasion, and HIF-1α protein expression and stability under hypoxia. These findings demonstrate that 15-LOX-1 expression loss in cancer cells promotes metastasis and that therapeutically targeting ubiquitous 15-LOX-1 loss in cancer cells has the potential to suppress metastasis.


Assuntos
Araquidonato 15-Lipoxigenase/biossíntese , Neoplasias do Colo/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Araquidonato 15-Lipoxigenase/genética , Hipóxia Celular/genética , Linhagem Celular Tumoral , Movimento Celular , Neoplasias do Colo/patologia , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Metástase Neoplásica , Fator A de Crescimento do Endotélio Vascular
10.
J Natl Cancer Inst ; 104(9): 709-16, 2012 May 02.
Artigo em Inglês | MEDLINE | ID: mdl-22472308

RESUMO

Expression of 15-lipoxygenase-1 (15-LOX-1) is decreased in many human cancers; however, the mechanistic significance of its decreased expression has been difficult to determine because its mouse homolog 12/15-LOX has opposing functions. We generated a mouse model in which expression of a human 15-LOX-1 transgene was targeted to the intestinal epithelium via the villin promoter. Targeted expression was confirmed by real-time reverse transcription-polymerase chain reaction and immunoblotting. When the 15-LOX-1 transgene was expressed in colonic epithelial cells of two independent mouse lines (B6 and FVB), azoxymethane-inducible colonic tumorigenesis was suppressed (mean number of tumors: wild type [WT] = 8.2, 15-LOX-1(+/-) = 4.91, 15-LOX-1(+/+) = 3.57; WT vs 15-LOX-1(+/-) two-sided P = .003, WT vs 15-LOX-1(+/+) two-sided P < .001; n = 10-14 mice per group). 15-LOX-1 transgene expression was always decreased in the tumors that did develop. In the presence of expression of the 15-LOX-1 transgene, expression of tumor necrosis factor alpha and its target inducible nitric oxide synthase were decreased and activation of nuclear factor-kappa B in colonic epithelial cells was inhibited.


Assuntos
Araquidonato 15-Lipoxigenase/genética , Araquidonato 15-Lipoxigenase/metabolismo , Colo/enzimologia , Neoplasias do Colo/enzimologia , Mucosa Intestinal/enzimologia , Transgenes , Animais , Azoximetano , Carcinógenos , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Neoplasias do Colo/induzido quimicamente , Modelos Animais de Doenças , Células Epiteliais/enzimologia , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Immunoblotting , Camundongos , Camundongos Transgênicos , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fator de Necrose Tumoral alfa/metabolismo , Regulação para Cima
11.
Cancer Prev Res (Phila) ; 4(12): 1961-72, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21881028

RESUMO

Loss of terminal cell differentiation promotes tumorigenesis. 15-Lipoxygenase-1 (15-LOX-1) contributes to terminal cell differentiation in normal cells. The mechanistic significance of 15-LOX-1 expression loss in human cancers to terminal cell differentiation suppression is unknown. In a screen of 128 cancer cell lines representing more than 20 types of human cancer, we found that 15-LOX-1 mRNA expression levels were markedly lower than levels in terminally differentiated cells. Relative expression levels of 15-LOX-1 (relative to the level in terminally differentiated primary normal human-derived bronchial epithelial cells) were lower in 79% of the screened cancer cell lines than relative expression levels of p16 (INK4A), which promotes terminal cell differentiation and is considered one of the most commonly lost tumor suppressor genes in cancer cells. 15-LOX-1 was expressed during terminal differentiation in three-dimensional air-liquid interface cultures, and 15-LOX-1 expression and terminal differentiation occurred in immortalized nontransformed bronchial epithelial but not in lung cancer cell lines. 15-LOX-1 expression levels were lower in human tumors than in paired normal lung epithelia. Short hairpin RNA-mediated downregulation of 15-LOX-1 in Caco-2 cells blocked enterocyte-like differentiation, disrupted tight junction formation, and blocked E-cadherin and ZO-1 localization to the cell wall membrane. 15-LOX-1 episomal expression in Caco-2 and HT-29 colon cancer cells induced differentiation. Our findings indicate that 15-LOX-1 downregulation in cancer cells is an important mechanism for terminal cell differentiation dysregulation and support the potential therapeutic utility of 15-LOX-1 reexpression to inhibit tumorigenesis.


Assuntos
Araquidonato 15-Lipoxigenase/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Diferenciação Celular , Neoplasias do Colo/patologia , Neoplasias Pulmonares/patologia , Fosfatase Alcalina/metabolismo , Araquidonato 15-Lipoxigenase/química , Araquidonato 15-Lipoxigenase/genética , Western Blotting , Brônquios/citologia , Brônquios/enzimologia , Caderinas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Membrana Celular/metabolismo , Células Cultivadas , Neoplasias do Colo/enzimologia , Imunofluorescência , Humanos , Técnicas Imunoenzimáticas , Pulmão/enzimologia , Pulmão/patologia , Neoplasias Pulmonares/enzimologia , RNA Mensageiro/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real
12.
J Natl Cancer Inst ; 101(10): 762-7, 2009 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-19436036

RESUMO

Peroxisome proliferator-activated receptor-delta (PPAR-delta) is overexpressed in human colon cancer, but its contribution to colonic tumorigenesis is controversial. We generated a mouse model in which PPAR-delta was genetically disrupted in colonic epithelial cells by targeted deletion of exon 4. Elimination of colon-specific PPAR-delta expression was confirmed by real-time reverse transcription-polymerase chain reaction (real-time RT-PCR), immunoblotting, and activity assays. Mice with and without targeted PPAR-delta genetic disruption (10-11 mice per group) were tested for incidence of azoxymethane-induced colon tumors. The effects of targeted PPAR-delta deletion on vascular endothelial growth factor expression were determined by real-time RT-PCR. Targeted PPAR-delta genetic disruption inhibited colonic carcinogenesis: Mice with PPAR-delta((-/-)) colons developed 98.5% fewer tumors than wild-type mice (PPAR-delta((-/-)) vs wild-type, mean = 0.1 tumors per mouse vs 6.6 tumors per mouse, difference = 6.5 tumors per mouse, 95% confidence interval = 4.9 to 8.0 tumors per mouse, P < .001, two-sided test). Increased expression of vascular endothelial growth factor in colon tumors vs normal colon was suppressed by loss of PPAR-delta expression. These findings indicate that PPAR-delta has a crucial role in promoting colonic tumorigenesis.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/terapia , Regulação Neoplásica da Expressão Gênica , PPAR delta/genética , Animais , Azoximetano , Linhagem Celular Tumoral , Neoplasias do Colo/induzido quimicamente , Modelos Animais de Doenças , Éxons , Deleção de Genes , Humanos , Camundongos , Camundongos Transgênicos , Modelos Genéticos , PPAR delta/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator A de Crescimento do Endotélio Vascular/biossíntese
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...