Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 63
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Gastroenterology ; 161(6): 1813-1829, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34606846

RESUMO

Chronic inflammation is a known risk factor for gastrointestinal cancer. The evidence that nonsteroidal anti-inflammatory drugs suppress the incidence, growth, and metastasis of gastrointestinal cancer supports the concept that a nonsteroidal anti-inflammatory drug target, cyclooxygenase, and its downstream bioactive lipid products may provide one of the links between inflammation and cancer. Preclinical studies have demonstrated that the cyclooxygenase-2-prostaglandin E2 pathway can promote gastrointestinal cancer development. Although the role of this pathway in cancer has been investigated extensively for 2 decades, only recent studies have described its effects on host defenses against transformed epithelial cells. Overcoming tumor-immune evasion remains one of the major challenges in cancer immunotherapy. This review summarizes the impacts of the cyclooxygenase-2-prostaglandin E2 pathway on gastrointestinal cancer development. Our focus was to highlight recent advances in our understanding of how this pathway induces tumor immune evasion.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Neoplasias Gastrointestinais/enzimologia , Mediadores da Inflamação/metabolismo , Evasão Tumoral , Microambiente Tumoral/imunologia , Animais , Antineoplásicos/uso terapêutico , Fibroblastos Associados a Câncer/enzimologia , Fibroblastos Associados a Câncer/imunologia , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , Neoplasias Gastrointestinais/tratamento farmacológico , Neoplasias Gastrointestinais/imunologia , Neoplasias Gastrointestinais/patologia , Humanos , Imunoterapia , Linfócitos do Interstício Tumoral/enzimologia , Linfócitos do Interstício Tumoral/imunologia , Transdução de Sinais , Evasão Tumoral/efeitos dos fármacos , Macrófagos Associados a Tumor/enzimologia , Macrófagos Associados a Tumor/imunologia
2.
Gastroenterology ; 158(4): 971-984.e10, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31734182

RESUMO

BACKGROUND & AIMS: Prostaglandin E2 (PGE2) promotes colorectal tumor formation and progression by unknown mechanisms. We sought to identify microRNAs (miRNAs) that might mediate the effects of PGE2 on colorectal cancer (CRC) development. METHODS: We incubated LS174T colorectal cancer cells with PGE2 or without (control) and used miRNA-sequencing technology to compare expression patterns of miRNAs. We knocked down levels of specific miRNAs or proteins in cells using small interfering RNAs or genome editing. Cells were analyzed by immunoblot, quantitative polymerase chain reaction, chromosome immunoprecipitation, cell invasion, and luciferase reporter assays; we measured gene expression, binding activity, cell migration and invasion, and transcriptional activity of transcription factors. NOD-scidIL-2Rg-/- mice were given injections of LS174T cells, and growth of primary tumors and numbers of liver and lung metastases were quantified and analyzed by histology. We used public databases to identify correlations in gene expression pattern with patient outcomes. RESULTS: We identified miRNA 675-5p (miR675-5p) as the miRNA most highly up-regulated by incubation of colorectal cancer cells with PGE2. PGE2 increased expression of miR675-5p by activating expression of Myc, via activation of protein kinase B, also known as (AKT), nuclear factor κB, and ß-catenin. PGE2 increased the invasive activities of cultured CRC cells. LS174T cells incubated with PGE2 formed more liver and lung metastases in mice than control LS174T cells. We identified a 3' untranslated region in the TP53 messenger RNA that bound miR675-5p; binding resulted in loss of the p53 protein. Expression of miR675-5p or its precursor RNA, H19, correlated with expression of cyclooxygenase-1 and cyclooxygenase-2 and shorter survival times of patients with CRC. CONCLUSIONS: We found that treatment of mice with PGE2 increased CRC cells invasive activity and ability to form liver and lung metastases. PGE2 down-regulates expression of p53 by increasing expression of miR675-5p, which binds to and prevents translation of TP53 messenger RNA. These findings provide insight into the mechanisms by which PGE2 promotes tumor development and progression. Strategies to target PGE2 might be developed for treatment of CRC.


Assuntos
Neoplasias Colorretais/metabolismo , Dinoprostona/genética , MicroRNAs/metabolismo , Metástase Neoplásica/genética , Proteína Supressora de Tumor p53/metabolismo , Ciclo-Oxigenase 1/metabolismo , Ciclo-Oxigenase 2/metabolismo , Humanos , RNA Longo não Codificante/metabolismo , Células Tumorais Cultivadas , Regulação para Cima/genética
3.
Proc Natl Acad Sci U S A ; 111(19): 7084-9, 2014 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-24763687

RESUMO

Although epidemiologic and experimental evidence strongly implicates chronic inflammation and dietary fats as risk factors for cancer, the mechanisms underlying their contribution to carcinogenesis are poorly understood. Here we present genetic evidence demonstrating that deletion of peroxisome proliferator-activated receptor δ (PPARδ) attenuates colonic inflammation and colitis-associated adenoma formation/growth. Importantly, PPARδ is required for dextran sodium sulfate induction of proinflammatory mediators, including chemokines, cytokines, COX-2, and prostaglandin E2 (PGE2), in vivo. We further show that activation of PPARδ induces COX-2 expression in colonic epithelial cells. COX-2-derived PGE2 stimulates macrophages to produce proinflammatory chemokines and cytokines that are responsible for recruitment of leukocytes from the circulation to local sites of inflammation. Our results suggest that PPARδ promotes colonic inflammation and colitis-associated tumor growth via the COX-2-derived PGE2 signaling axis that mediates cross-talk between tumor epithelial cells and macrophages.


Assuntos
Adenoma/metabolismo , Colite/metabolismo , Neoplasias Colorretais/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Adenoma/patologia , Animais , Comunicação Celular/fisiologia , Linhagem Celular Tumoral , Colite/induzido quimicamente , Colite/patologia , Neoplasias Colorretais/patologia , Ciclo-Oxigenase 2/metabolismo , Sulfato de Dextrana/toxicidade , Gorduras na Dieta/metabolismo , Dinoprostona/metabolismo , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Humanos , Macrófagos/citologia , Macrófagos/metabolismo , Masculino , Camundongos , Camundongos Knockout , Monócitos/citologia , Monócitos/metabolismo , Receptores Citoplasmáticos e Nucleares/genética
4.
Gastroenterology ; 149(7): 1884-1895.e4, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26261008

RESUMO

BACKGROUND & AIMS: Inflammation may contribute to the formation, maintenance, and expansion of cancer stem cells (CSCs), which have the capacity for self-renewal, differentiation, and resistance to cytotoxic agents. We investigated the effects of the inflammatory mediator prostaglandin E2 (PGE2) on colorectal CSC development and metastasis in mice and the correlation between levels of PGE2 and CSC markers in human colorectal cancer (CRC) specimens. METHODS: Colorectal carcinoma specimens and matched normal tissues were collected from patients at the Mayo Clinic (Scottsdale, AZ) and analyzed by mass spectrometry and quantitative polymerase chain reaction. Human primary CRC cells and mouse tumor cells were isolated using microbeads or flow cytometry and analyzed for sphere-formation and by flow cytometry assays. LS-174T cells were sorted by flow cytometry (for CD133(+)CD44(+) and CD133(-)CD44(-) cells) and also used in these assays. NOD-scidIL-2Rγ(-/-) (NSG) mice were given cecal or subcutaneous injections of LS-174T or human primary CRC cells. Apc(Min/+) mice and NSG mice with orthotopic cecal tumors were given vehicle (controls), PGE2, celecoxib, and/or Ono-AE3-208. PGE2 downstream signaling pathways were knocked down with small hairpin RNAs, expressed from lentiviral vectors in LS-174T cells, or blocked with inhibitors in human primary CRC cells. RESULTS: Levels of PGE2 correlated with colonic CSC markers (CD133, CD44, LRG5, and SOX2 messenger RNAs) in human colorectal carcinoma samples. Administration of PGE2 to Apc(Min/+) mice increased tumor stem cells and tumor burden, compared with controls. NSG mice given PGE2 had increased numbers of cecal CSCs and liver metastases compared with controls after intracecal injection of LS-174T or human primary CRC cells. Alternatively, celecoxib, an inhibitor of prostaglandin-endoperoxide synthase 2, reduced polyp numbers in Apc(Min/+) mice, liver metastasis in NSG mice with orthotopic tumors, and numbers of CSCs in Apc(Min/+) and NSG mice. Inhibitors or knockdown of PGE2 receptor 4 (EP4), phosphoinositide 3-kinase (PI3K) p85α, extracellular signal-regulated kinase 1 (ERK1), or nuclear factor (NF)-κB reduced PGE2-induced sphere formation and expansion of LS-174T and/or human primary CRC cells. Knockdown of ERK1 or PI3K p85α also attenuated PGE2-induced activation of NF-κB in LS-174T cells. An EP4 antagonist reduced the ability of PGE2 to induce CSC expansion in orthotopic tumors and to accelerate the formation of liver metastases. Knockdown experiments showed that NF-κB was required for PGE2 induction of CSCs and metastasis in mice. CONCLUSIONS: PGE2 induces CSC expansion by activating NF-κB, via EP4-PI3K and EP4-mitogen-activated protein kinase signaling, and promotes the formation of liver metastases in mice. The PGE2 signaling pathway therefore might be targeted therapeutically to slow CSC expansion and colorectal cancer progression.


Assuntos
Carcinoma/metabolismo , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/metabolismo , Dinoprostona/metabolismo , Mediadores da Inflamação/metabolismo , Células-Tronco Neoplásicas/metabolismo , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma/genética , Carcinoma/prevenção & controle , Carcinoma/secundário , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/prevenção & controle , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/administração & dosagem , Regulação Neoplásica da Expressão Gênica , Genes APC , Xenoenxertos , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/prevenção & controle , Neoplasias Hepáticas/secundário , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Fosfatidilinositol 3-Quinase/genética , Fosfatidilinositol 3-Quinase/metabolismo , Interferência de RNA , Receptores de Prostaglandina E Subtipo EP4/genética , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Transdução de Sinais , Transfecção , Carga Tumoral , Células Tumorais Cultivadas
5.
Carcinogenesis ; 36(10): 1085-93, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26354776

RESUMO

Chronic inflammation contributes to cancer development via multiple mechanisms. One potential mechanism is that chronic inflammation can generate an immunosuppressive microenvironment that allows advantages for tumor formation and progression. The immunosuppressive environment in certain chronic inflammatory diseases and solid cancers is characterized by accumulation of proinflammatory mediators, infiltration of immune suppressor cells and activation of immune checkpoint pathways in effector T cells. In this review, we highlight recent advances in our understanding of how immunosuppression contributes to cancer and how proinflammatory mediators induce the immunosuppressive microenvironment via induction of immunosuppressive cells and activation of immune checkpoint pathways.


Assuntos
Inflamação/imunologia , Melanoma/imunologia , Linfócitos T/imunologia , Microambiente Tumoral/imunologia , Doença Crônica , Humanos , Tolerância Imunológica , Terapia de Imunossupressão , Inflamação/patologia , Melanoma/patologia , Linfócitos T/patologia
6.
Annu Rev Med ; 64: 131-44, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23020877

RESUMO

A large body of evidence indicates that genetic mutations, epigenetic changes, chronic inflammation, diet, and lifestyle are key risk factors for colorectal cancer (CRC). Prevention of CRC has long been considered a plausible approach for the population and individuals at high risk for developing this disease. A significant effort has been made in the development of novel drugs for both prevention and treatment over the past two decades. This review highlights recent advances in our understanding of the role of nonsteroidal anti-inflammatory drugs in CRC prevention and adjuvant treatment. Moreover, we focus on the molecular mechanisms underlying the antitumor effects of these drugs in CRC. The knowledge of how anti-inflammatory agents inhibit cancer formation and progression may provide a rationale for the development of more effective chemopreventive and chemotherapeutic agents with less toxicity.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Neoplasias Colorretais/prevenção & controle , Neoplasias Colorretais/epidemiologia , Saúde Global , Humanos , Morbidade/tendências , Prognóstico
7.
Invest New Drugs ; 32(6): 1105-1112, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-25085205

RESUMO

Nonsteroidal anti-inflammatory drugs (NSAIDs) and selective cyclooxygenase-2 (COX-2) inhibitors (COXIBs) can reduce the risk of developing colorectal cancer (CRC) and are being considered for use as adjuvant therapy for treatment of CRC patients. However, long-term use of most NSAIDs, except aspirin, increases cardiovascular risk, hampering use of these drugs in CRC prevention and possibly for treatment. CG100649 is a new member of the COXIB family, which is proposed to inhibit both COX-2 and carbonic anhydrase-I/-II (CA-I/-II) activity. Using mouse models, we show here that CG100649 inhibits premalignant and malignant colorectal lesions in mouse models, partly through inhibiting tumor cell proliferation. These pre-clinical findings suggest a need for further exploration of CG100649 for CRC prevention and treatment. The long-term safety profile of CG100649, particularly regarding its effect on cardiovascular risk, is yet to be determined.


Assuntos
Adenoma/tratamento farmacológico , Antineoplásicos/uso terapêutico , Carcinoma/tratamento farmacológico , Neoplasias Colorretais/tratamento farmacológico , Inibidores de Ciclo-Oxigenase 2/uso terapêutico , Furanos/uso terapêutico , Sulfonamidas/uso terapêutico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Dinoprostona/metabolismo , Furanos/farmacologia , Humanos , Camundongos , Sulfonamidas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Cancer Res Commun ; 3(8): 1486-1500, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37559947

RESUMO

While the role of prostaglandin E2 (PGE2) in promoting malignant progression is well established, how to optimally block the activity of PGE2 signaling remains to be demonstrated. Clinical trials with prostaglandin pathway targeted agents have shown activity but without sufficient significance or dose-limiting toxicities that have prevented approval. PGE2 signals through four receptors (EP1-4) to modulate tumor progression. EP2 and EP4 signaling exacerbates tumor pathology and is immunosuppressive through potentiating cAMP production. EP1 and EP3 signaling has the opposite effect through increasing IP3 and decreasing cAMP. Using available small-molecule antagonists of single EP receptors, the cyclooxygenase-2 (COX-2) inhibitor celecoxib, or a novel dual EP2/EP4 antagonist generated in this investigation, we tested which approach to block PGE2 signaling optimally restored immunologic activity in mouse and human immune cells and antitumor activity in syngeneic, spontaneous, and xenograft tumor models. We found that dual antagonism of EP2 and EP4 together significantly enhanced the activation of PGE2-suppressed mouse and human monocytes and CD8+ T cells in vitro as compared with single EP antagonists. CD8+ T-cell activation was dampened by single EP1 and EP3 antagonists. Dual EP2/EP4 PGE2 receptor antagonists increased tumor microenvironment lymphocyte infiltration and significantly reduced disease burden in multiple tumor models, including in the adenomatous polyposis coli (APC)min+/- spontaneous colorectal tumor model, compared with celecoxib. These results support a hypothesis that redundancy of EP2 and EP4 receptor signaling necessitates a therapeutic strategy of dual blockade of EP2 and EP4. Here we describe TPST-1495, a first-in-class orally available small-molecule dual EP2/EP4 antagonist. Significance: Prostaglandin (PGE2) drives tumor progression but the pathway has not been effectively drugged. We demonstrate significantly enhanced immunologic potency and antitumor activity through blockade of EP2 and EP4 PGE2 receptor signaling together with a single molecule.


Assuntos
Neoplasias , Prostaglandinas , Humanos , Animais , Camundongos , Dinoprostona/metabolismo , Receptores de Prostaglandina E Subtipo EP2/metabolismo , Celecoxib/farmacologia , Linfócitos T CD8-Positivos/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo , Inibidores de Ciclo-Oxigenase 2 , Microambiente Tumoral
9.
J Exp Med ; 203(4): 941-51, 2006 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-16567391

RESUMO

Chronic inflammation is a well-known risk factor for cancer. Proinflammatory mediators such as prostaglandin E2 (PGE2) promote colorectal tumor growth by stimulating angiogenesis, cell invasion, and cell growth, and inhibiting apoptosis. Molecules that regulate tumor-associated angiogenesis provide promising therapeutic targets for treatment of colorectal cancer (CRC) as indicated by the recent development of the novel anti-angiogenic agent bevacizumab (Avastin). However, use of this drug only prolongs survival by several months, highlighting the importance of finding more effective treatment regimens. We report here that PGE2 induces expression of CXCL1 (growth-regulated oncogene alpha), a pro-angiogenic chemokine, in human CRC cells. More importantly, CXCL1 released from carcinoma cells induces microvascular endothelial cell migration and tube formation in vitro. Furthermore, PGE2 promotes tumor growth in vivo by induction of CXCL1 expression, which results in increased tumor microvessel formation. These results have potential clinical significance because we found that CXCL1 expression correlates with PGE2 levels in human CRCs. Collectively, our findings show for the first time that CXCL1 is regulated by PGE2 and indicate that CXCL1 inhibitors should be evaluated further as potential anti-angiogenic agents for treatment of CRC.


Assuntos
Quimiocinas CXC/fisiologia , Neoplasias Colorretais/irrigação sanguínea , Dinoprostona/fisiologia , Peptídeos e Proteínas de Sinalização Intercelular/fisiologia , Neovascularização Patológica/imunologia , Neovascularização Patológica/metabolismo , Adenoma/irrigação sanguínea , Adenoma/patologia , Animais , Células CACO-2 , Linhagem Celular Tumoral , Movimento Celular/imunologia , Quimiocina CXCL1 , Quimiocinas CXC/biossíntese , Quimiocinas CXC/genética , Quimiocinas CXC/metabolismo , Neoplasias Colorretais/patologia , Endotélio Vascular/metabolismo , Endotélio Vascular/patologia , Receptores ErbB/fisiologia , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Peptídeos e Proteínas de Sinalização Intercelular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos SCID , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Receptores de Interleucina-8B/biossíntese , Receptores de Interleucina-8B/genética
10.
Gastroenterology ; 141(2): 553-64, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21704622

RESUMO

BACKGROUND & AIMS: Colonization of gastric mucosa by Helicobacter pylori leads to epithelial hyperproliferation, which increases the risk for gastric adenocarcinoma. One H pylori virulence locus associated with cancer risk, cag, encodes a secretion system that transports effectors into host cells and leads to aberrant activation of ß-catenin and p120-catenin (p120). Peroxisome proliferator-activated receptor (PPAR)δ is a ligand-activated transcription factor that affects oncogenesis in conjunction with ß-catenin. We used a carcinogenic H pylori strain to define the role of microbial virulence constituents and PPARδ in regulating epithelial responses that mediate development of adenocarcinoma. METHODS: Gastric epithelial cells or colonies were co-cultured with the H pylori cag(+) strain 7.13 or cagE(-), cagA(-), soluble lytic transglycosylase(-), or cagA(-)/soluble lytic transglycosylase(-) mutants. Levels of PPARδ and cyclin E1 were determined by real-time, reverse-transcription polymerase chain reaction, immunoblot analysis, or immunofluorescence microscopy; proliferation was measured in 3-dimensional culture. PPARδ and Ki67 expression were determined by immunohistochemical analysis of human biopsies and rodent gastric mucosa. RESULTS: H pylori induced ß-catenin- and p120-dependent expression and activation of PPARδ in gastric epithelial cells, which were mediated by the cag secretion system substrates CagA and peptidoglycan. H pylori stimulated proliferation in vitro, which required PPARδ-mediated activation of cyclin E1; H pylori did not induce expression of cyclin E1 in a genetic model of PPARδ deficiency. PPARδ expression and proliferation in rodent and human gastric tissue was selectively induced by cag(+) strains and PPARδ levels normalized after eradication of H pylori. CONCLUSIONS: The H pylori cag secretion system activates ß-catenin, p120, and PPARδ, which promote gastric epithelial cell proliferation via activation of cyclin E1. PPARδ might contribute to gastric adenocarcinoma development in humans.


Assuntos
Adenocarcinoma/microbiologia , Células Epiteliais/metabolismo , Infecções por Helicobacter/metabolismo , Helicobacter pylori/metabolismo , PPAR delta/metabolismo , Neoplasias Gástricas/microbiologia , Adenocarcinoma/patologia , Animais , Antígenos de Bactérias/genética , Antígenos de Bactérias/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Cateninas/metabolismo , Proliferação de Células , Transformação Celular Neoplásica , Células Cultivadas , Ciclina E/metabolismo , Células Epiteliais/microbiologia , Mucosa Gástrica/metabolismo , Mucosa Gástrica/patologia , Gerbillinae , Helicobacter pylori/genética , Humanos , Antígeno Ki-67/metabolismo , Proteínas Oncogênicas/metabolismo , Transdução de Sinais , Neoplasias Gástricas/patologia , beta Catenina/metabolismo , delta Catenina
11.
Cancer Cell ; 6(3): 285-95, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15380519

RESUMO

Cyclooxygenase-derived prostaglandin E(2) (PGE(2)) is the predominant prostanoid found in most colorectal cancers (CRC) and is known to promote colon carcinoma growth and invasion. However, the key downstream signaling pathways necessary for PGE(2)-induced intestinal carcinogenesis are unclear. Here we report that PGE(2) indirectly transactivates PPARdelta through PI3K/Akt signaling, which promotes cell survival and intestinal adenoma formation. We also found that PGE(2) treatment of Apc(min) mice dramatically increased intestinal adenoma burden, which was negated in Apc(min) mice lacking PPARdelta. We demonstrate that PPARdelta is a focal point of crosstalk between the prostaglandin and Wnt signaling pathways which results in a shift from cell death to cell survival, leading to increased tumor growth.


Assuntos
Adenoma/metabolismo , Neoplasias Colorretais/metabolismo , Dinoprostona/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Animais , Sobrevivência Celular , Cromonas/farmacologia , Dinoprostona/farmacologia , Genes APC , Humanos , Masculino , Camundongos , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-akt , Transfecção , Células Tumorais Cultivadas
12.
Cancer Prev Res (Phila) ; 15(5): 285-296, 2022 05 03.
Artigo em Inglês | MEDLINE | ID: mdl-35121582

RESUMO

The mechanisms underlying the regulation of a checkpoint receptor, PD-1, in tumor-infiltrating immune cells during the development of colorectal cancer are not fully understood. Here we demonstrate that COX-2-derived PGE2, an inflammatory mediator and tumor promoter, induces PD-1 expression by enhancing NFκB's binding to the PD-1 promoter via an EP4-PI3K-Akt signaling pathway in both CD8+ T cells and macrophages. Moreover, PGE2 suppresses CD8+ T-cell proliferation and cytotoxicity against tumor cells and impairs macrophage phagocytosis of cancer cells via an EP4-PI3K-Akt-NFκB-PD-1 signaling pathway. In contrast, inhibiting the COX-2-PGE2-EP4 pathway increases intestinal CD8+ T-cell activation and proliferation and enhances intestinal macrophage phagocytosis of carcinoma cells accompanied by reduction of PD-1 expression in intestinal CD8+ T cells and macrophages in ApcMin/+ mice. PD-1 expression correlates well with COX-2 levels in human colorectal cancer specimens. Both elevated PD-1 and COX-2 are associated with poorer overall survival in patients with colorectal cancer. Our results uncover a novel role of PGE2 in tumor immune evasion. They may provide the rationale for developing new therapeutic approaches to subvert this process by targeting immune checkpoint pathways using EP4 antagonists. In addition, our findings reveal a novel mechanism explaining how NSAIDs reduce colorectal cancer risk by suppressing tumor immune evasion. PREVENTION RELEVANCE: These findings provide a potential explanation underlying the chemopreventive effect of NSAIDs on reducing colorectal cancer incidence during premalignancy and provide a rationale for developing EP4 antagonists for colorectal cancer prevention and treatment. Simply targeting PGE2 signaling alone may be efficacious in colorectal cancer prevention and treatment, avoiding side effects associated with NSAIDs.


Assuntos
Adenoma , Neoplasias Colorretais , Animais , Anti-Inflamatórios não Esteroides , Neoplasias Colorretais/patologia , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Humanos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Receptor de Morte Celular Programada 1 , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Prostaglandina E Subtipo EP4/metabolismo
13.
Nat Med ; 10(3): 245-7, 2004 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-14758356

RESUMO

We treated Apc(min) mice, which are predisposed to intestinal polyposis, with a selective synthetic agonist of peroxisome proliferator-activated receptor-delta (PPAR-delta). Exposure of Apc(min) mice to the PPAR-delta ligand GW501516 resulted in a significant increase in the number and size of intestinal polyps. The most prominent effect was on polyp size; mice treated with the PPAR-delta activator had a fivefold increase in the number of polyps larger than 2 mm. Our results implicate PPAR-delta in the regulation of intestinal adenoma growth.


Assuntos
Adenoma/patologia , Neoplasias Intestinais/patologia , Pólipos Intestinais/patologia , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores Citoplasmáticos e Nucleares/metabolismo , Tiazóis/farmacologia , Fatores de Transcrição/agonistas , Fatores de Transcrição/metabolismo , Adenoma/metabolismo , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Genes APC , Humanos , Neoplasias Intestinais/metabolismo , Pólipos Intestinais/metabolismo , Ligantes , Camundongos , Tiazóis/metabolismo
14.
Nat Med ; 10(10): 1074-80, 2004 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-15378054

RESUMO

Ectopic pregnancy is a major reproductive health issue. Although other underlying causes remain largely unknown, one cause of ectopic pregnancy is embryo retention in the fallopian tube. Here we show that genetic or pharmacologic silencing of cannabinoid receptor CB1 causes retention of a large number of embryos in the mouse oviduct, eventually leading to pregnancy failure. This is reversed by isoproterenol, a beta-adrenergic receptor agonist. Impaired oviductal embryo transport is also observed in wild-type mice treated with methanandamide. Collectively, the results suggest that aberrant cannabinoid signaling impedes coordinated oviductal smooth muscle contraction and relaxation crucial to normal oviductal embryo transport. Colocalization of CB1 and beta2-adrenergic receptors in the oviduct muscularis implies that a basal endocannabinoid tone in collaboration with adrenergic receptors coordinates oviductal motility for normal journey of embryos into the uterus. Besides uncovering a new regulatory mechanism, this study could be clinically relevant to ectopic pregnancy.


Assuntos
Agonistas Adrenérgicos beta/farmacologia , Isoproterenol/farmacologia , Norepinefrina/metabolismo , Transporte do Óvulo/efeitos dos fármacos , Receptor CB1 de Canabinoide/metabolismo , Transdução de Sinais/fisiologia , Animais , Ácidos Araquidônicos , Southern Blotting , Transferência Embrionária , Tubas Uterinas/metabolismo , Tubas Uterinas/fisiopatologia , Feminino , Camundongos , Modelos Genéticos , Contração Muscular/fisiologia , Transporte do Óvulo/fisiologia , Gravidez , Receptor CB1 de Canabinoide/deficiência , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
Oncogene ; 40(41): 5984-5992, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34385594

RESUMO

PD-L1 expression is elevated in various human cancers, including colorectal cancer. High levels of PD-L1 expressed on tumor epithelial cells are one of the potential mechanisms by which tumor cells become resistant to immune attack. However, PD-L1 regulation in tumor cells is not fully understood. Here we demonstrate that mutations in the adenomatous polyposis coli (APC) gene lead to colonic epithelial cell resistance to CD8+ T cell cytotoxicity by induction of PD-L1 expression. Mechanistically, this occurs as a result of the ß-catenin/TCF4 complex binding to the PD-L1 promoter, leading to increased transcription. Our findings not only reveal a novel mechanism by which APC mutations induce tumor immune evasion via an immune checkpoint pathway but also pave the way for developing ß-catenin or TCF4 inhibitors as possible new options for immune checkpoint inhibition.


Assuntos
Proteína da Polipose Adenomatosa do Colo/genética , Antígeno B7-H1/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/imunologia , Proteína da Polipose Adenomatosa do Colo/imunologia , Animais , Antígeno B7-H1/biossíntese , Antígeno B7-H1/genética , Linfócitos T CD8-Positivos/imunologia , Neoplasias Colorretais/tratamento farmacológico , Técnicas de Silenciamento de Genes , Células HEK293 , Células HT29 , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Evasão Tumoral/genética
16.
Cancer Res ; 67(11): 5285-92, 2007 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-17545608

RESUMO

The underlying causes of epithelial ovarian cancer (EOC) are unclear, and treatment options for patients with advanced disease are limited. There is evidence that the use of nonsteroidal anti-inflammatory drugs is associated with decreased risk of developing EOC. Nonsteroidal anti-inflammatory drugs inhibit cyclooxygenase (COX)-1 and COX-2, which catalyze prostaglandin biosynthesis. We previously showed that mouse and human EOCs have increased levels of COX-1, but not COX-2, and a COX-1-selective inhibitor, SC-560, attenuates prostaglandin production and tumor growth. However, the downstream targets of COX-1 signaling in EOC are not yet known. To address this question, we evaluated peroxisome proliferator-activated receptor delta (PPARdelta) expression and function in EOC. We found that EOC cells express high levels of PPARdelta, and neutralizing PPARdelta function reduces tumor growth in vivo. More interestingly, aspirin, a nonsteroidal anti-inflammatory drug that preferentially inhibits COX-1, compromises PPARdelta function and cell growth by inhibiting extracellular signal-regulated kinases 1/2, members of the mitogen-activated protein kinase family. Our study, for the first time, shows that whereas PPARdelta can be a target of COX-1, extracellular signal-regulated kinase is a potential target of PPARdelta. The ability of aspirin to inhibit EOC growth in vivo is an exciting finding because of its low cost, lack of cardiovascular side effects, and availability.


Assuntos
Ciclo-Oxigenase 1/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias Ovarianas/metabolismo , PPAR delta/metabolismo , Animais , Anti-Inflamatórios não Esteroides/farmacologia , Aspirina/farmacologia , Processos de Crescimento Celular/efeitos dos fármacos , Processos de Crescimento Celular/fisiologia , Linhagem Celular Tumoral , Inibidores de Ciclo-Oxigenase/farmacologia , Ativação Enzimática , Células Epiteliais/patologia , Epoprostenol/metabolismo , Feminino , Humanos , Masculino , Camundongos , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/enzimologia , Neoplasias Ovarianas/patologia , PPAR delta/antagonistas & inibidores , PPAR delta/biossíntese , Pirazóis/farmacologia , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Cancer Res ; 79(17): 4480-4490, 2019 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-31239272

RESUMO

The nuclear hormone receptor peroxisome proliferator-activated receptor delta (PPARδ) is a ligand-dependent transcription factor involved in fatty acid metabolism, obesity, wound healing, inflammation, and cancer. Although PPARδ has been shown to promote intestinal adenoma formation and growth, the molecular mechanisms underlying the contribution of PPARδ to colorectal cancer remain unclear. Here, we demonstrate that activation of PPARδ induces expansion of colonic cancer stem cells (CSC) and promotes colorectal cancer liver metastasis by binding to the Nanog promoter and enhancing Nanog expression. Moreover, PPARδ mediated the effect of a high-fat diet in promoting liver metastasis and induction of colonic CSC expansion. Our findings uncover a novel role of dietary fats in colorectal cancer metastasis and reveal novel mechanisms underlying PPARδ-mediated induction of CSCs and those responsible for the contribution of dietary fats to colorectal cancer progression. These findings may provide a rationale for developing PPARδ antagonists to therapeutically target CSCs in colorectal cancer. SIGNIFICANCE: These findings show that PPARδ contributes to colorectal cancer metastasis by expanding the CSC population, indicating that antagonists that target PPARδ may be beneficial in treating colorectal cancer.


Assuntos
Neoplasias Colorretais/patologia , Gorduras na Dieta/efeitos adversos , PPAR delta/metabolismo , Animais , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/metabolismo , Dieta Hiperlipídica/efeitos adversos , Células HCT116 , Humanos , Neoplasias Hepáticas Experimentais/secundário , Masculino , Camundongos Endogâmicos NOD , Camundongos Mutantes , Proteína Homeobox Nanog/genética , Células-Tronco Neoplásicas/patologia , PPAR delta/agonistas , Regiões Promotoras Genéticas , Tiazóis/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
19.
Am J Respir Cell Mol Biol ; 39(6): 689-96, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18566335

RESUMO

Peroxisome proliferator-activated receptor beta/delta (PPARbeta/delta) is a ligand-binding inducible transcriptional factor linked to carcinogenesis. Important functions of PPARbeta/delta were demonstrated in series of human epithelial cancers; however, its role in lung cancer remains controversial. We investigated the differential expression level and localization of PPARbeta/delta in tumors and adjacent normal lung tissue, and the effect of PPARbeta/delta activation on lung cancer cell proliferation and apoptosis. PPARbeta/delta was expressed in all studied human non-small cell lung cancers, and strong PPARbeta/delta immunoreactivity was observed in epithelial cells of more than 75% of studied lung tumors. PPARbeta/delta expression was consistently limited to the cancer cells in tumor tissue, while in adjacent normal lung tissue it was limited predominantly to the mononuclear cells. We found that ligand-binding activation of PPARbeta/delta stimulates cell proliferation (an effect that was blocked by a dominant-negative construct of PPARbeta/delta), stimulates anchorage-independent cell growth, and inhibits apoptosis in lung cancer cell lines. Importantly, the activation of PPARbeta/delta induces Akt phosphorylation correlated with up-regulation of PDK1, down-regulation of PTEN, and increased expression of Bcl-xL and COX-2. These findings indicate that PPARbeta/delta exerts proliferative and anti-apoptotic effects via PI3K/Akt1 and COX-2 pathways. In conclusion, PPARbeta/delta is strongly expressed in the majority of lung cancers, and its activation induces proliferative and survival response in non-small cell lung cancer.


Assuntos
Neoplasias Pulmonares/metabolismo , PPAR delta/metabolismo , PPAR beta/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Apoptose , Adesão Celular , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Ciclo-Oxigenase 2/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Ligantes , Pulmão/metabolismo , Pulmão/patologia , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , PPAR delta/antagonistas & inibidores , PPAR beta/antagonistas & inibidores , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Análise Serial de Tecidos
20.
J Clin Invest ; 128(7): 2732-2742, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29733297

RESUMO

Chronic inflammation is a risk factor for gastrointestinal cancer and other diseases. Most studies have focused on cytokines and chemokines as mediators connecting chronic inflammation to cancer, whereas the involvement of lipid mediators, including prostanoids, has not been extensively investigated. Prostanoids are among the earliest signaling molecules released in response to inflammation. Multiple lines of evidence suggest that prostanoids are involved in gastrointestinal cancer. In this Review, we discuss how prostanoids impact gastrointestinal cancer development. In particular, we highlight recent advances in our understanding of how prostaglandin E2 induces the immunosuppressive microenvironment in gastrointestinal cancers.


Assuntos
Neoplasias Gastrointestinais/etiologia , Neoplasias Gastrointestinais/metabolismo , Prostaglandinas/metabolismo , Animais , Carcinógenos/metabolismo , Dinoprostona/imunologia , Dinoprostona/metabolismo , Neoplasias Gastrointestinais/imunologia , Humanos , Tolerância Imunológica , Mediadores da Inflamação/imunologia , Mediadores da Inflamação/metabolismo , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Redes e Vias Metabólicas , Modelos Biológicos , Prostaglandinas/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Microambiente Tumoral/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA