Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cancer Sci ; 115(2): 401-411, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38041233

RESUMEN

Desmoid tumors (DTs), also called desmoid-type fibromatoses, are locally aggressive tumors of mesenchymal origin. In the present study, we developed a novel mouse model of DTs by inducing a local mutation in the Ctnnb1 gene, encoding ß-catenin in PDGFRA-positive stromal cells, by subcutaneous injection of 4-hydroxy-tamoxifen. Tumors in this model resembled histologically clinical samples from DT patients and showed strong phosphorylation of nuclear SMAD2. Knockout of SMAD4 in the model significantly suppressed tumor growth. Proteomic analysis revealed that SMAD4 knockout reduced the level of Cysteine-and-Glycine-Rich Protein 2 (CSRP2) in DTs, and treatment of DT-derived cells with a TGF-ß receptor inhibitor reduced CSRP2 RNA levels. Knockdown of CSRP2 in DT cells significantly suppressed their proliferation. These results indicate that the TGF-ß/CSRP2 axis is a potential therapeutic target for DTs downstream of TGF-ß signaling.


Asunto(s)
Fibromatosis Agresiva , Animales , Humanos , Ratones , beta Catenina/genética , beta Catenina/metabolismo , Fibromatosis Agresiva/genética , Fibromatosis Agresiva/patología , Proteínas con Dominio LIM/genética , Ratones Noqueados , Proteínas Musculares/metabolismo , Proteínas Nucleares/genética , Proteómica , Factor de Crecimiento Transformador beta/metabolismo , Regulación hacia Arriba
2.
Nat Commun ; 14(1): 6246, 2023 10 06.
Artículo en Inglés | MEDLINE | ID: mdl-37803016

RESUMEN

Cancer cachexia is a complex metabolic disorder accounting for ~20% of cancer-related deaths, yet its metabolic landscape remains unexplored. Here, we report a decrease in B vitamin-related liver enzymes as a hallmark of systemic metabolic changes occurring in cancer cachexia. Metabolomics of multiple mouse models highlights cachexia-associated reductions of niacin, vitamin B6, and a glycine-related subset of one-carbon (C1) metabolites in the liver. Integration of proteomics and metabolomics reveals that liver enzymes related to niacin, vitamin B6, and glycine-related C1 enzymes dependent on B vitamins decrease linearly with their associated metabolites, likely reflecting stoichiometric cofactor-enzyme interactions. The decrease of B vitamin-related enzymes is also found to depend on protein abundance and cofactor subtype. These metabolic/proteomic changes and decreased protein malonylation, another cachexia feature identified by protein post-translational modification analysis, are reflected in blood samples from mouse models and gastric cancer patients with cachexia, underscoring the clinical relevance of our findings.


Asunto(s)
Niacina , Neoplasias Gástricas , Complejo Vitamínico B , Ratones , Animales , Humanos , Caquexia/etiología , Caquexia/metabolismo , Proteómica , Piridoxina , Vitamina B 6 , Hígado/metabolismo , Glicina/metabolismo
4.
Cancer Gene Ther ; 28(12): 1339-1352, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-33414520

RESUMEN

The prognosis of glioblastoma remains poor despite intensive research efforts. Glioblastoma stem cells (GSCs) contribute to tumorigenesis, invasive capacity, and therapy resistance. Leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5), a stem cell marker, is involved in the maintenance of GSCs, although the properties of Lgr5-positive GSCs remain unclear. Here, the Sleeping-Beauty transposon-induced glioblastoma model was used in Lgr5-GFP knock-in mice identify GFP-positive cells in neurosphere cultures from mouse glioblastoma tissues. Global gene expression analysis showed that Gli2 was highly expressed in GFP-positive GSCs. Gli2 knockdown using lentiviral-mediated shRNA downregulated Hedgehog-related and Wnt signaling pathway-related genes, including Lgr5; suppressed tumor cell proliferation and invasion capacity; and induced apoptosis. Pharmacological Gli inhibition with GANT61 suppressed tumor cell proliferation. Silencing Gli2 suppressed the tumorigenicity of GSCs in an orthotopic transplantation model in vivo. These findings suggest that Gli2 affects the Hedgehog and Wnt pathways and plays an important role in GSC maintenance, suggesting Gli2 as a therapeutic target for glioblastoma treatment.


Asunto(s)
Neoplasias Encefálicas/genética , Perfilación de la Expresión Génica/métodos , Glioblastoma/genética , Proteína Gli2 con Dedos de Zinc/metabolismo , Animales , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Proliferación Celular , Modelos Animales de Enfermedad , Glioblastoma/patología , Humanos , Ratones , Pronóstico
5.
Cancer Sci ; 112(4): 1614-1623, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33506575

RESUMEN

We previously reported that ROR1 is a crucial downstream gene for the TTF-1/NKX2-1 lineage-survival oncogene in lung adenocarcinoma, while others have found altered expression of ROR1 in multiple cancer types. Accumulated evidence therefore indicates ROR1 as an attractive molecular target, though it has yet to be determined whether targeting Ror1 can inhibit tumor development and growth in vivo. To this end, genetically engineered mice carrying homozygously floxed Ror1 alleles and an SP-C promoter-driven human mutant EGFR transgene were generated. Ror1 ablation resulted in marked retardation of tumor development and progression in association with reduced malignant characteristics and significantly better survival. Interestingly, gene set enrichment analysis identified a hypoxia-induced gene set (HALLMARK_HYPOXIA) as most significantly downregulated by Ror1 ablation in vivo, which led to findings showing that ROR1 knockdown diminished HIF-1α expression under normoxia and clearly hampered HIF-1α induction in response to hypoxia in human lung adenocarcinoma cell lines. The present results directly demonstrate the importance of Ror1 for in vivo development and progression of lung adenocarcinoma, and also identify Ror1 as a novel regulator of Hif-1α. Thus, a future study aimed at the development of a novel therapeutic targeting ROR1 for treatment of solid tumors such as seen in lung cancer, which are frequently accompanied with a hypoxic tumor microenvironment, is warranted.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Pulmonares/genética , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/genética , Adenocarcinoma del Pulmón/patología , Animales , Línea Celular Tumoral , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Hipoxia/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Oncogenes/genética , Transducción de Señal/genética , Factor Nuclear Tiroideo 1/genética , Microambiente Tumoral/genética
6.
Oncogene ; 40(2): 408-420, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33177648

RESUMEN

Although the Wnt/ß-catenin pathway plays a central role in the carcinogenesis and maintenance of colorectal cancer (CRC), attempts to target the pathway itself have not been very successful. MyD88, an adaptor protein of the TLR/IL-1ß signaling, has been implicated in the integrity of the intestines as well as in their tumorigenesis. In this study, we aimed to clarify the mechanisms by which epithelial MyD88 contributes to intestinal tumor formation and to address whether MyD88 can be a therapeutic target of CRC. Conditional knockout of MyD88 in intestinal epithelial cells (IECs) reduced tumor formation in Apc+/Δ716 mice, accompanied by decreased proliferation and enhanced apoptosis of tumor epithelial cells. Mechanistically, the MyD88 loss caused inactivation of the JNK-mTORC1, NF-κB, and Wnt/ß-catenin pathways in tumor cells. Induction of MyD88 knockout in the intestinal tumor-derived organoids, but not in the normal IEC-derived organoids, induced apoptosis and reduced their growth. Treatment with the MyD88 inhibitor ST2825 also suppressed the growth of the intestinal tumor-derived organoids. Knockdown of MYD88 in human CRC cell lines with mutations in APC or CTNNB1 induced apoptosis and reduced their proliferation as well. These results indicate that MyD88 loss is synthetic lethal with mutational activation of the Wnt/ß-catenin signaling in intestinal tumor epithelial cells. Inhibition of MyD88 signaling can thus be a novel therapeutic strategy for familial adenomatous polyposis (FAP) as well as for colorectal cancer harboring mutations in the Wnt/ß-catenin signaling.


Asunto(s)
Mucosa Intestinal/patología , Neoplasias Intestinales/patología , Factor 88 de Diferenciación Mieloide/fisiología , Mutaciones Letales Sintéticas , Proteínas Wnt/genética , beta Catenina/genética , Animales , Apoptosis , Proliferación Celular , Células Cultivadas , Femenino , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/genética , Neoplasias Intestinales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
7.
Cancer Sci ; 110(8): 2520-2528, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31215118

RESUMEN

Iodothyronine deiodinase 2 (DIO2) converts the prohormone thyroxine (T4) to bioactive T3 in peripheral tissues and thereby regulates local thyroid hormone (TH) levels. Although epidemiologic studies suggest the contribution of TH to the progression of colorectal cancer (CRC), the role of DIO2 in CRC remains elusive. Here we show that Dio2 is highly expressed in intestinal polyps of ApcΔ716 mice, a mouse model of familial adenomatous polyposis and early stage sporadic CRC. Laser capture microdissection and in situ hybridization analysis show almost exclusive expression of Dio2 in the stroma of ApcΔ716 polyps in the proximity of the COX-2-positive areas. Treatment with iopanoic acid, a deiodinase inhibitor, or chemical thyroidectomy suppresses tumor formation in ApcΔ716 mice, accompanied by reduced tumor cell proliferation and angiogenesis. Dio2 expression in ApcΔ716 polyps is strongly suppressed by treatment with the COX-2 inhibitor meloxicam. Analysis of The Cancer Genome Atlas data shows upregulation of DIO2 in CRC clinical samples and a close association of its expression pattern with the stromal component, consistently with almost exclusive expression of DIO2 in the stroma of human CRC as revealed by in situ hybridization. These results indicate essential roles of stromal DIO2 and thyroid hormone signaling in promoting the growth of intestinal tumors.


Asunto(s)
Proliferación Celular/fisiología , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Yoduro Peroxidasa/metabolismo , Poliposis Adenomatosa del Colon/tratamiento farmacológico , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Animales , Proliferación Celular/efectos de los fármacos , Neoplasias Colorrectales/tratamiento farmacológico , Ciclooxigenasa 2/metabolismo , Inhibidores de la Ciclooxigenasa 2/farmacología , Modelos Animales de Enfermedad , Humanos , Pólipos Intestinales/tratamiento farmacológico , Pólipos Intestinales/metabolismo , Pólipos Intestinales/patología , Ratones , Ratones Noqueados , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Hormonas Tiroideas/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/fisiología , Yodotironina Deyodinasa Tipo II
8.
Oncogene ; 38(11): 1966-1978, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30401981

RESUMEN

Malignant mesothelioma (MM) constitutes a very aggressive tumor that is caused by asbestos exposure after long latency. The NF2 tumor suppressor gene is mutated in 40-50% of MM; moreover, one of its downstream signaling cascades, the Hippo signaling pathway, is also frequently inactivated in MM cells. Although the YAP transcriptional coactivator, which is regulated by the Hippo pathway, can function as a pro-oncogenic protein, the role of TAZ, a paralog of YAP, in MM cells has not yet been clarified. Here, we show that TAZ is expressed and underphosphorylated (activated) in the majority of MM cells compared to immortalized mesothelial cells. ShRNA-mediated TAZ knockdown highly suppressed cell proliferation, anchorage-independent growth, cell motility, and invasion in MM cells harboring activated TAZ. Conversely, transduction of an activated form of TAZ in immortalized mesothelial cells enhanced these in vitro phenotypes and conferred tumorigenicity in vivo. Microarray analysis determined that activated TAZ most significantly enhanced the transcription of genes related to "cytokine-cytokine receptor interaction." Among selected cytokines, we found that IL-1 signaling activation plays a major role in proliferation in TAZ-activated MM cells. Both IL1B knockdown and an IL-1 receptor antagonist significantly suppressed malignant phenotypes of immortalized mesothelial cells and MM cells with activated TAZ. Overall, these results indicate an oncogenic role for TAZ in MMs via transcriptional induction of distinct pro-oncogenic genes including cytokines. Among these, IL-1 signaling appears as one of the most important cascades, thus potentially serving as a target pathway in MM cells harboring Hippo pathway inactivation.


Asunto(s)
Transformación Celular Neoplásica/genética , Citocinas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/genética , Mesotelioma/genética , Proteínas Serina-Treonina Quinasas/genética , Factores de Transcripción/genética , Animales , Línea Celular Tumoral , Citocinas/metabolismo , Epitelio/metabolismo , Epitelio/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Vía de Señalización Hippo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patología , Mesotelioma/metabolismo , Mesotelioma/patología , Mesotelioma Maligno , Ratones , Ratones Desnudos , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/genética , Transactivadores , Factores de Transcripción/metabolismo , Activación Transcripcional , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ
9.
Proc Natl Acad Sci U S A ; 114(37): E7697-E7706, 2017 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-28847964

RESUMEN

Cancer cells alter their metabolism for the production of precursors of macromolecules. However, the control mechanisms underlying this reprogramming are poorly understood. Here we show that metabolic reprogramming of colorectal cancer is caused chiefly by aberrant MYC expression. Multiomics-based analyses of paired normal and tumor tissues from 275 patients with colorectal cancer revealed that metabolic alterations occur at the adenoma stage of carcinogenesis, in a manner not associated with specific gene mutations involved in colorectal carcinogenesis. MYC expression induced at least 215 metabolic reactions by changing the expression levels of 121 metabolic genes and 39 transporter genes. Further, MYC negatively regulated the expression of genes involved in mitochondrial biogenesis and maintenance but positively regulated genes involved in DNA and histone methylation. Knockdown of MYC in colorectal cancer cells reset the altered metabolism and suppressed cell growth. Moreover, inhibition of MYC target pyrimidine synthesis genes such as CAD, UMPS, and CTPS blocked cell growth, and thus are potential targets for colorectal cancer therapy.


Asunto(s)
Adenoma/metabolismo , Neoplasias Colorrectales/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Adenoma/genética , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Proliferación Celular/fisiología , Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Femenino , Genes myc , Humanos , Masculino , Metabolómica/métodos , Ratones , Proteínas Proto-Oncogénicas c-myc/genética , Pirimidinas/biosíntesis , Transcriptoma
10.
Curr Top Microbiol Immunol ; 407: 153-189, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28550454

RESUMEN

The PI3K/AKT/mTOR pathway is frequently activated in various human cancers and has been considered a promising therapeutic target. Many of the positive regulators of the PI3K/AKT/mTOR axis, including the catalytic (p110α) and regulatory (p85α), of class IA PI3K, AKT, RHEB, mTOR, and eIF4E, possess oncogenic potentials, as demonstrated by transformation assays in vitro and by genetically engineered mouse models in vivo. Genetic evidences also indicate their roles in malignancies induced by activation of the upstream oncoproteins including receptor tyrosine kinases and RAS and those induced by the loss of the negative regulators of the PI3K/AKT/mTOR pathway such as PTEN, TSC1/2, LKB1, and PIPP. Possible mechanisms by which the PI3K/AKT/mTOR axis contributes to oncogenic transformation include stimulation of proliferation, survival, metabolic reprogramming, and invasion/metastasis, as well as suppression of autophagy and senescence. These phenotypic changes are mediated by eIF4E-induced translation of a subset of mRNAs and by other downstream effectors of mTORC1 including S6K, HIF-1α, PGC-1α, SREBP, and ULK1 complex.


Asunto(s)
Transformación Celular Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Animales , Humanos
11.
Cancer Sci ; 106(6): 692-699, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25855137

RESUMEN

Extracellular signal-regulated kinase is an MAPK that is most closely associated with cell proliferation, and the MEK/ERK signaling pathway is implicated in various human cancers. Although epidermal growth factor receptor, KRAS, and BRAF are considered major targets for colon cancer treatment, the precise roles of the MEK/ERK pathway, one of their major downstream effectors, during colon cancer development remain to be determined. Using Apc(Δ716) mice, a mouse model of familial adenomatous polyposis and early-stage sporadic colon cancer formation, we show that MEK/ERK signaling is activated not only in adenoma epithelial cells, but also in tumor stromal cells including fibroblasts and vascular endothelial cells. Eight-week treatment of Apc(Δ716) mice with trametinib, a small-molecule MEK inhibitor, significantly reduced the number of polyps in the large size class, accompanied by reduced angiogenesis and tumor cell proliferation. Trametinib treatment reduced the COX-2 level in Apc(Δ716) tumors in vivo and in primary culture of intestinal fibroblasts in vitro. Antibody array analysis revealed that trametinib and the COX-2 inhibitor rofecoxib both reduced the level of CCL2, a chemokine known to be essential for the growth of Apc mutant polyps, in intestinal fibroblasts in vitro. Consistently, trametinib treatment reduced the Ccl2 mRNA level in Apc(Δ716) tumors in vivo. These results suggest that MEK/ERK signaling plays key roles in intestinal adenoma formation in Apc(Δ716) mice, at least in part, through COX-2 induction in tumor stromal cells.


Asunto(s)
Antineoplásicos/farmacología , Ciclooxigenasa 2/análisis , Genes APC/fisiología , Pólipos Intestinales/tratamiento farmacológico , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridonas/farmacología , Pirimidinonas/farmacología , Animales , Quimiocina CCL2/análisis , Quimiocina CCL2/genética , Modelos Animales de Enfermedad , Femenino , Pólipos Intestinales/enzimología , Masculino , Ratones
12.
Clin Exp Metastasis ; 31(8): 977-89, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25326065

RESUMEN

To understand colon cancer metastasis, we earlier analyzed a mouse model that developed liver metastasis of cancer cells disseminated from the spleen. We suggested that CCR1(+) bone marrow (BM)-derived cells are recruited to the microenvironment of disseminated colon cancer cells, and produce metalloproteinases MMP9 and MMP2, helping metastatic colonization. In the present study, we have examined these myeloid cells expressing CCR1 and/or MMPs in detail. To this end, we have established bacterial artificial chromosome (BAC)-based transgenic mouse lines in which membrane-targeted Venus fluorescent protein (mVenus) was expressed under the control of Ccr1 gene promoter. Then, myeloid cells obtained from the BM and liver metastatic foci were analyzed by the combination of flow cytometry and cytology/immunohistochemistry, in situ RNA hybridization, or quantitative RT-PCR. We have found four distinct types of myeloid cells recruited to the metastatic foci; neutrophils, eosinophils, monocytes and fibrocytes. These cell types exhibited distinct expression patterns for CCR1, MMP2 and MMP9. Namely, neutrophils found in the early phase of cancer cell dissemination expressed CCR1 exclusively and MMP9 preferentially, whereas fibrocytes accumulated in later phase expressed MMP2 exclusively. Either genetic inactivation of Ccr1 or antibody-mediated neutrophil depletion reduced subsequent recruitment of fibrocytes. The recruitment of CCR1(+) neutrophils in early phase of colon cancer dissemination appears to cause that of fibrocytes in late phase. These results implicate the key role of CCR1 in colon cancer metastasis in this mouse model, and explain why both MMP9 and MMP2 are essential as genetically demonstrated previously. The results also suggest relevant mechanisms in humans.


Asunto(s)
Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Neoplasias Hepáticas/secundario , Células Mieloides/patología , Receptores CCR1/fisiología , Microambiente Tumoral , Animales , Western Blotting , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Ciclo Celular , Movimiento Celular , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Citometría de Flujo , Humanos , Técnicas para Inmunoenzimas , Hibridación in Situ , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Células Mieloides/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Tumorales Cultivadas
13.
Gastroenterology ; 145(5): 1064-1075.e11, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23891973

RESUMEN

BACKGROUND & AIMS: Loss of the tumor suppressor SMAD4 correlates with progression of colorectal cancer (CRC). In mice, colon tumors that express CCL9 recruit CCR1(+) myeloid cells, which facilitate tumor invasion and metastasis by secreting matrix metalloproteinase 9. METHODS: We used human CRC cell lines to investigate the ability of SMAD4 to regulate expression of CCL15, a human ortholog of mouse CCL9. We used immunohistochemistry to compare levels of CCL15 and other proteins in 141 samples of human liver metastases. RESULTS: In human CRC cell lines, knockdown of SMAD4 increased CCL15 expression, and overexpression of SMAD4 decreased it. SMAD4 bound directly to the promoter region of the CCL15 gene to negatively regulate its expression; transforming growth factor-ß increased binding of SMAD4 to the CCL15 promoter and transcriptional repression. In livers of nude mice, SMAD4-deficient human CRC cells up-regulated CCL15 to recruit CCR1(+) cells and promote metastasis. In human tumor samples, there was a strong inverse correlation between levels of CCL15 and SMAD4; metastases that expressed CCL15 contained 3-fold more CCR1(+) cells than those without CCL15. Patients with CCL15-expressing metastases had significantly shorter times of disease-free survival than those with CCL15-negative metastases. CCR1(+) cells in the metastases expressed the myeloid cell markers CD11b and myeloperoxidase, and also matrix metalloproteinase 9. CONCLUSIONS: In human CRC cells, loss of SMAD4 leads to up-regulation of CCL15 expression. Human liver metastases that express CCL15 contain higher numbers CCR1(+) cells; patients with these metastases have shorter times of disease-free survival. Reagents designed to block CCL15 recruitment of CCR1(+) cells could prevent metastasis of CRC to liver.


Asunto(s)
Adenocarcinoma/metabolismo , Quimiocinas CC/metabolismo , Neoplasias Colorrectales/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas Inflamatorias de Macrófagos/metabolismo , Células Mieloides/patología , Receptores CCR1/metabolismo , Proteína Smad4/deficiencia , Adenocarcinoma/mortalidad , Adenocarcinoma/secundario , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígeno CD11b/metabolismo , Línea Celular Tumoral , Neoplasias Colorrectales/mortalidad , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/fisiología , Xenoinjertos , Humanos , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/secundario , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Desnudos , Persona de Mediana Edad , Células Mieloides/metabolismo , Metástasis de la Neoplasia/fisiopatología , Metástasis de la Neoplasia/prevención & control , Peroxidasa/metabolismo , Estudios Retrospectivos , Proteína Smad4/efectos de los fármacos , Proteína Smad4/genética , Tasa de Supervivencia
14.
Int J Clin Oncol ; 16(5): 464-72, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21847533

RESUMEN

Colorectal cancer is the second most common cancer, and is the third leading cause of cancer-related death in Japan. The majority of these deaths is attributable to liver metastasis. Recent studies have provided increasing evidence that the chemokine-chemokine receptor system is a potential mechanism of tumor metastasis via multiple complementary actions: (a) by promoting cancer cell migration, invasion, survival and angiogenesis; and (b) by recruiting distal stromal cells (i.e., myeloid bone marrow-derived cells) to indirectly facilitate tumor invasion and metastasis. Here, we discuss recent preclinical and clinical data supporting the view that chemokine pathways are potential therapeutic targets for liver metastasis of colorectal cancer.


Asunto(s)
Quimiocinas/metabolismo , Neoplasias Colorrectales/patología , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Receptores de Quimiocina/metabolismo , Movimiento Celular/genética , Quimiocinas/antagonistas & inhibidores , Quimiocinas/genética , Humanos , Neoplasias Hepáticas/tratamiento farmacológico , Terapia Molecular Dirigida , Células Mieloides/metabolismo , Células Mieloides/patología , Invasividad Neoplásica , Neovascularización Patológica/metabolismo , Receptores de Quimiocina/antagonistas & inhibidores , Receptores de Quimiocina/genética , Microambiente Tumoral
15.
Gastroenterology ; 140(5): 1556-63.e6, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21320501

RESUMEN

BACKGROUND & AIMS: Signaling by the mammalian target of rapamycin complex 1 (mTORC1) has been implicated in various human cancers. mTORC1 signaling is activated in intestinal tumors of adenomatous polyposis coli (Apc(Δ716)) mice, a model of familial adenomatous polyposis; in these mice, the mTORC1 inhibitor RAD001 can block tumor formation. However, the precise mechanism of mTORC1 signaling in intestinal tumors is not clear. We investigated whether c-Jun-NH(2) terminal kinase (JNK) is involved in the mTORC1 activation. METHODS: We investigated the effects of an inhibitor and an activator of JNK, as well as small interfering RNA against JNK, on mTORC1 in Apc(Δ716) mice and colon cancer cell lines. We also determined the role of JNK in mTORC1 signaling using in vitro kinase assays. RESULTS: JNK was activated in intestinal polyps of Apc((Δ716) mice); the JNK inhibitor SP600125 significantly suppressed tumor formation. In colorectal cancer cell lines, the JNK activator anisomycin activated mTORC1, whereas SP600125 or small interfering RNAs against JNK suppressed signaling. Importantly, JNK stimulated the mTORC1 kinase activity in vitro, through direct phosphorylation of Raptor at serine 863. CONCLUSIONS: JNK is required for activation of mTORC1 in intestinal tumor cells. JNK inhibitors might be developed as therapeutics or to prevent development of intestinal tumors.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Regulación Neoplásica de la Expresión Génica , MAP Quinasa Quinasa 4/metabolismo , Proteínas/genética , ARN Neoplásico/genética , Poliposis Adenomatosa del Colon/metabolismo , Poliposis Adenomatosa del Colon/patología , Animales , Western Blotting , Línea Celular Tumoral , Femenino , Inmunohistoquímica , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Noqueados , Complejos Multiproteicos , Neoplasias Experimentales , Proteínas/metabolismo , ARN Neoplásico/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Serina-Treonina Quinasas TOR
16.
Proc Natl Acad Sci U S A ; 107(29): 13063-8, 2010 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-20616008

RESUMEN

Recent reports have suggested critical roles of myeloid cells in tumor invasion and metastasis, although these findings have not led to therapeutics. Using a mouse model for liver dissemination, we show that mouse and human colon cancer cells secrete CC-chemokine ligands CCL9 and CCL15, respectively, and recruit CD34(+) Gr-1(-) immature myeloid cells (iMCs). They express CCL9/15 receptor CCR1 and produce matrix metalloproteinases MMP2 and MMP9. Lack of the Ccr1, Mmp2, or Mmp9 gene in the host dramatically suppresses outgrowths of disseminated tumors in the liver. Importantly, CCR1 antagonist BL5923 blocks the iMC accumulation and metastatic colonization and significantly prolongs the survival of tumor-bearing mice. These results suggest that CCR1 antagonists can provide antimetastatic therapies for patients with disseminated colon cancer in the liver.


Asunto(s)
Diferenciación Celular , Neoplasias del Colon/patología , Neoplasias Hepáticas/secundario , Células Mieloides/patología , Receptores CCR1/metabolismo , Animales , Quimiocinas CC/metabolismo , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Humanos , Ligandos , Hígado/metabolismo , Hígado/patología , Neoplasias Hepáticas/enzimología , Neoplasias Hepáticas/patología , Proteínas Inflamatorias de Macrófagos/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Células Mieloides/metabolismo , Receptores CCR1/antagonistas & inhibidores , Transducción de Señal , Análisis de Supervivencia
17.
Cell Cycle ; 8(22): 3684-7, 2009 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-19855161

RESUMEN

Activation of the mTORC1 pathway has been implicated in many types of cancer, and several mTORC1 inhibitors are currently under clinical trials for treating various cancer patients. Notably, Temsirolimus has recently been approved for treatment of advanced renal cell carcinoma. However, the role of mTORC1 pathway in colorectal tumorigenesis remains largely unknown. We have recently found that the mTORC1 pathway is activated in intestinal adenomas of Apc mutant mice, accompanied by an elevated level of mTOR protein, and that treatment with RAD001, an mTORC1 inhibitor, suppresses the growth of these polyps. Our results suggest an important role of mTORC1 pathway in colorectal cancer, as well as a therapeutic possibility for mTORC1 inhibitors in its treatment.


Asunto(s)
Adenoma/metabolismo , Neoplasias Intestinales/metabolismo , Modelos Biológicos , Transducción de Señal/fisiología , Factores de Transcripción/metabolismo , Adenoma/tratamiento farmacológico , Animales , Everolimus , Humanos , Neoplasias Intestinales/tratamiento farmacológico , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Complejos Multiproteicos , Proteínas , Sirolimus/análogos & derivados , Sirolimus/farmacología , Serina-Treonina Quinasas TOR , Factores de Transcripción/antagonistas & inhibidores , Proteínas Wnt/metabolismo
18.
Proc Natl Acad Sci U S A ; 105(36): 13544-9, 2008 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-18768809

RESUMEN

The mammalian target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth via mTOR complex 1 (mTORC1), whose activation has been implicated in many human cancers. However, mTORC1's status in gastrointestinal tumors has not been characterized thoroughly. We have found that the mTORC1 pathway is activated with increased expression of the mTOR protein in intestinal polyps of the Apc(Delta716) heterozygous mutant mouse, a model for human familial adenomatous polyposis. An 8-week treatment with RAD001 (everolimus) suppressed the mTORC1 activity in these polyps and inhibited proliferation of the adenoma cells as well as tumor angiogenesis, which significantly reduced not only the number of polyps but also their size. beta-Catenin knockdown in the colon cancer cell lines reduced the mTOR level and thereby inhibited the mTORC1 signaling. These results suggest that the Wnt signaling contributes to mTORC1 activation through the increased level of mTOR and that the activation plays important roles in the intestinal polyp formation and growth. Indeed, long-term RAD001 treatment significantly reduced mortality of the Apc(Delta716) mice. Thus, we propose that the mTOR inhibitors may be efficacious for therapy and prevention of colonic adenomas and cancers with Wnt signaling activation.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Pólipos Intestinales/metabolismo , Pólipos Intestinales/prevención & control , Transducción de Señal , Factores de Transcripción/metabolismo , Adenoma/irrigación sanguínea , Adenoma/tratamiento farmacológico , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Animales , Proliferación Celular/efectos de los fármacos , Everolimus , Femenino , Pólipos Intestinales/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Transgénicos , Complejos Multiproteicos , Proteínas , Transducción de Señal/efectos de los fármacos , Sirolimus/análogos & derivados , Sirolimus/farmacología , Sirolimus/uso terapéutico , Tasa de Supervivencia , Serina-Treonina Quinasas TOR , Proteínas Wnt/metabolismo
19.
Pharmacol Ther ; 98(1): 109-27, 2003 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-12667890

RESUMEN

Pharmacological agents, such as histamine H(2) receptor antagonists and acid pump inhibitors, are now the most frequently used treatment for such acid-related diseases as gastroduodenal ulcers and reflux esophagitis. Based on increased understanding of the precise mechanisms of gastric acid secretion at the level of receptors, enzymes, and cytoplasmic signal transduction systems, further possibilities exist for the development of effective antisecretory pharmacotherapy. Gastrin CCK(2) receptor antagonists and locally active agents appear to represent promising therapies for the future. Development of gene targeting techniques has allowed production of genetically engineered transgenic and knockout mice. Such genetic technology has increased the investigative power for pharmacotherapy for not only antisecretory agents, but also treatment of mucosal diseases, such as atrophy, hyperplasia, and cancer. Elucidation of the origin of gastric parietal cells also represents an interesting investigative target that should allow a better understanding of not only acid-related diseases, but also the evolution of the stomach as an acid-secreting organ.


Asunto(s)
Antiulcerosos/uso terapéutico , Ácido Gástrico/metabolismo , Úlcera Péptica/tratamiento farmacológico , Animales , Ácido Gástrico/fisiología , Humanos , Ratones , Ratones Noqueados/genética , Células Parietales Gástricas/fisiología , Úlcera Péptica/fisiopatología
20.
Gastroenterology ; 125(6): 1774-84, 2003 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-14724830

RESUMEN

BACKGROUND & AIMS: The physiologic significance of the M(3) muscarinic receptor is unclear due to an absence of specific ligand. In the present study, M(3) receptor knockout (KO) mice were used to elucidate the role of M(3) receptors in gastric acid secretion and gastric mucosal integrity. METHODS: M(3) KO versus wild-type mice aged 1 month to 2 years were included. Gastric acid secretion was assessed by both direct intragastric pH measurement and pylorus ligation. Serum gastrin and gastric mucosal histamine levels were determined by radioimmunoassay and enzyme-linked immunosorbent assay, respectively. Morphologic analysis was performed by both immunohistochemistry and transmission electron microscopy. RESULTS: Fasted M(3) KO mice exhibited higher intragastric pH, lower acid output after pylorus ligation, a lower proportion of active parietal cells, and higher serum gastrin levels than fasted wild-type mice. Acid secretion in response to carbachol, histamine, gastrin 17, and 2-deoxy-D-glucose was impaired in the mutant mice. Although carbachol was still able to cause approximately 30% acid output in M(3) KO mice, the acid secretion was inhibited by pirenzepine or famotidine. Despite remarkable hypergastrinemia in M(3) KO mice, there were no trophic responses in the oxyntic mucosa with respect to the mucosal thickness, proliferation rate, and numbers of parietal and enterochromaffin-like cells. Cholecystokinin type 2 receptor antagonist YM022 was without the effect in M(3) KO mice. CONCLUSIONS: The present study shows that M(3) receptors are essential for basal acid secretion, a fully acid secretory response to histamine and gastrin, and the trophic responses of oxyntic mucosa to gastrin.


Asunto(s)
Mucosa Gástrica/metabolismo , Gastrinas/sangre , Receptor Muscarínico M3/fisiología , Animales , Benzodiazepinas/farmacología , Calcio/metabolismo , División Celular , Células Enterocromafines/metabolismo , Femenino , Ácido Gástrico/metabolismo , Mucosa Gástrica/patología , Histamina/análisis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...