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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gastroenterology ; 163(5): 1228-1241, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-35870513

RESUMEN

BACKGROUND & AIMS: Mechanisms contributing to the onset and progression of Barrett's (BE)-associated esophageal adenocarcinoma (EAC) remain elusive. Here, we interrogated the major signaling pathways deregulated early in the development of Barrett's neoplasia. METHODS: Whole-transcriptome RNA sequencing analysis was performed in primary BE, EAC, normal esophageal squamous, and gastric biopsy tissues (n = 89). Select pathway components were confirmed by quantitative polymerase chain reaction in an independent cohort of premalignant and malignant biopsy tissues (n = 885). Functional impact of selected pathway was interrogated using transcriptomic, proteomic, and pharmacogenetic analyses in mammalian esophageal organotypic and patient-derived BE/EAC cell line models, in vitro and/or in vivo. RESULTS: The vast majority of primary BE/EAC tissues and cell line models showed hyperactivation of EphB2 signaling. Transcriptomic/proteomic analyses identified EphB2 as an endogenous binding partner of MYC binding protein 2, and an upstream regulator of c-MYC. Knockdown of EphB2 significantly impeded the viability/proliferation of EAC and BE cells in vitro/in vivo. Activation of EphB2 in normal esophageal squamous 3-dimensional organotypes disrupted epithelial maturation and promoted columnar differentiation programs, notably including MYC. EphB2 and MYC showed selective induction in esophageal submucosal glands with acinar ductal metaplasia, and in a porcine model of BE-like esophageal submucosal gland spheroids. Clinically approved inhibitors of MEK, a protein kinase that regulates MYC, effectively suppressed EAC tumor growth in vivo. CONCLUSIONS: The EphB2 signaling is frequently hyperactivated across the BE-EAC continuum. EphB2 is an upstream regulator of MYC, and activation of EphB2-MYC axis likely precedes BE development. Targeting EphB2/MYC could be a promising therapeutic strategy for this often refractory and aggressive cancer.


Asunto(s)
Esófago de Barrett , Carcinoma de Células Escamosas , Neoplasias Esofágicas , Porcinos , Animales , Esófago de Barrett/patología , Efrina-B2/genética , Proteómica , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas/patología , Proto-Oncogenes , Proteínas Tirosina Quinasas/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Mamíferos/genética
2.
Haematologica ; 103(6): 1054-1064, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29472361

RESUMEN

Hematopoietic stem cell transplantation following myeloablative chemotherapy is a curative treatment for many hematopoietic malignancies. However, profound granulocytopenia during the interval between transplantation and marrow recovery exposes recipients to risks of fatal infection, a significant source of transplant-associated morbidity and mortality. We have previously described the discovery of a small molecule, SW033291, that potently inhibits the prostaglandin degrading enzyme 15-PGDH, increases bone marrow prostaglandin E2, and accelerates hematopoietic recovery following murine transplant. Here we describe the efficacy of (+)-SW209415, a second-generation 15-PGDH inhibitor, in an expanded range of models relevant to human transplantation. (+)-SW209415 is 10,000-fold more soluble, providing the potential for intravenous delivery, while maintaining potency in inhibiting 15-PGDH, increasing in vivo prostaglandin E2, and accelerating hematopoietic regeneration following transplantation. In additional models, (+)-SW209415: (i) demonstrated synergy with granulocyte colony-stimulating factor, the current standard of care; (ii) maintained efficacy as transplant cell dose was escalated; (iii) maintained efficacy when transplant donors and recipients were aged; and (iv) potentiated homing in xenotransplants using human hematopoietic stem cells. (+)-SW209415 showed no adverse effects, no potentiation of in vivo growth of human myeloma and leukemia xenografts, and, on chronic high-dose administration, no toxicity as assessed by weight, blood counts and serum chemistry. These studies provide independent chemical confirmation of the activity of 15-PGDH inhibitors in potentiating hematopoietic recovery, extend the range of models in which inhibiting 15-PGDH demonstrates activity, allay concerns regarding potential for adverse effects from increasing prostaglandin E2, and thereby, advance 15-PGDH as a therapeutic target for potentiating hematopoietic stem cell transplantation.


Asunto(s)
Antineoplásicos/farmacología , Inhibidores Enzimáticos/farmacología , Supervivencia de Injerto/efectos de los fármacos , Factor Estimulante de Colonias de Granulocitos/farmacología , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Adulto , Factores de Edad , Animales , Trasplante de Médula Ósea , Femenino , Trasplante de Células Madre Hematopoyéticas , Xenoinjertos , Humanos , Masculino , Ratones
3.
Carcinogenesis ; 36(2): 291-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25503930

RESUMEN

Non-steroidal anti-inflammatory drugs prevent colorectal cancer by inhibiting cyclooxygenase (COX) enzymes that synthesize tumor-promoting prostaglandins. 15-hydroxyprostaglandin dehydrogenase (15-PGDH) is a tumor suppressor that degrades tumor-promoting prostaglandins. Murine knockout of 15-PGDH increases susceptibility to azoxymethane-induced colon tumors. It also renders these mice resistant to celecoxib, a selective inhibitor of inducible COX-2 during colon neoplasia. Similarly, humans with low colonic 15-PGDH are also resistant to colon adenoma prevention with celecoxib. Here, we used aspirin and sulindac, which inhibit both COX-1 and COX-2, in order to determine if these broader COX inhibitors can prevent colon tumors in 15-PGDH knockout (KO) mice. Unlike celecoxib, sulindac proved highly effective in colon tumor prevention of 15-PGDH KO mice. Significantly, however, aspirin demonstrated no effect on colon tumor incidence in either 15-PGDH wild-type or KO mice, despite a comparable reduction in colonic mucosal Prostaglandin E2 (PGE2) levels by both sulindac and aspirin. Notably, colon tumor prevention activity by sulindac was accompanied by a marked induction of lymphoid aggregates and proximal colonic inflammatory mass lesions, a side effect seen to a lesser degree with celecoxib, but not with aspirin. These findings suggest that sulindac may be the most effective agent for colon cancer prevention in humans with low 15-PGDH, but its use may also be associated with inflammatory lesions in the colon.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/prevención & control , Hidroxiprostaglandina Deshidrogenasas/genética , Sulindac/farmacología , Animales , Antineoplásicos/farmacología , Aspirina/farmacología , Azoximetano , Carcinógenos , Celecoxib , Quimioprevención , Neoplasias del Colon/inducido químicamente , Ciclooxigenasa 1/efectos de los fármacos , Ciclooxigenasa 2/efectos de los fármacos , Inhibidores de la Ciclooxigenasa 2/farmacología , Dinoprostona/metabolismo , Inflamación/inmunología , Mucosa Intestinal/patología , Proteínas de la Membrana/efectos de los fármacos , Ratones , Ratones Noqueados , Pirazoles/farmacología , Sulfonamidas/farmacología
4.
Int J Gynecol Cancer ; 25(3): 484-92, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25594147

RESUMEN

PURPOSE: The aim of this study was to evaluate the tolerability and efficacy of poly(ADP-ribose) polymerase (PARP) inhibition by veliparib during cytotoxic topotecan administration with filgrastim or pegfilgrastim neutrophil support in women with persistent or recurrent uterine cervix cancer. EXPERIMENTAL DESIGN: This phase I-II trial examined twice-daily oral veliparib (10 mg) given during once-daily intravenous topotecan (0.6 mg/m²) on days 1 to 5 of each treatment cycle. Cycles were repeated every 21 days until disease progression or until toxicity prohibited further therapy. Toxicity and objective response rate were primary endpoints. RESULTS: Twenty-seven women were enrolled. Frequently reported grade 3 or higher treatment-related toxicities were anemia (59%), thrombocytopenia (44%), leukopenia (22%), and neutropenia (19%). There were 2 partial responses (7% [90% confidence interval, 1%-22%]). Four patients had a disease progression date more than 6 months after the start of veliparib-topotecan therapy. Patients with low immunohistochemical expression (0-1+) of PARP-1 in their primary uterine cervix cancer were more likely to have a longer progression-free interval (hazard ratio, 0.25; P = 0.02) and survival (hazard ratio, 0.12; P = 0.005) after veliparib-topotecan therapy. CONCLUSIONS: Clinical activity of a veliparib-topotecan combination was minimal in women with persistent or recurrent uterine cervix cancer. Women whose uterine cervix cancers express PARP-1 at low levels may benefit preferentially from PARP inhibitors combined with cytotoxic therapies, suggesting further study of PARP expression as an integral triage biomarker.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Carcinoma/tratamiento farmacológico , Recurrencia Local de Neoplasia/tratamiento farmacológico , Neoplasias del Cuello Uterino/tratamiento farmacológico , Adulto , Anciano , Anemia/inducido químicamente , Bencimidazoles/administración & dosificación , Bencimidazoles/efectos adversos , Carcinoma/química , Proteínas de Ciclo Celular/análisis , Progresión de la Enfermedad , Femenino , Filgrastim/uso terapéutico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Persona de Mediana Edad , Recurrencia Local de Neoplasia/química , Neutropenia/inducido químicamente , Neutropenia/prevención & control , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/análisis , Polietilenglicoles , Proteínas Recombinantes/uso terapéutico , Ribonucleótido Reductasas/análisis , Trombocitopenia/inducido químicamente , Topotecan/administración & dosificación , Topotecan/efectos adversos , Neoplasias del Cuello Uterino/química
5.
Proc Natl Acad Sci U S A ; 108(49): E1312-20, 2011 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-22042863

RESUMEN

Ablation of the kinases Mst1 and Mst2, orthologs of the Drosophila antiproliferative kinase Hippo, from mouse intestinal epithelium caused marked expansion of an undifferentiated stem cell compartment and loss of secretory cells throughout the small and large intestine. Although median survival of mice lacking intestinal Mst1/Mst2 is 13 wk, adenomas of the distal colon are common by this age. Diminished phosphorylation, enhanced abundance, and nuclear localization of the transcriptional coactivator Yes-associated protein 1 (Yap1) is evident in Mst1/Mst2-deficient intestinal epithelium, as is strong activation of ß-catenin and Notch signaling. Although biallelic deletion of Yap1 from intestinal epithelium has little effect on intestinal development, inactivation of a single Yap1 allele reduces Yap1 polypeptide abundance to nearly wild-type levels and, despite the continued Yap hypophosphorylation and preferential nuclear localization, normalizes epithelial structure. Thus, supraphysiologic Yap polypeptide levels are necessary to drive intestinal stem cell proliferation. Yap is overexpressed in 68 of 71 human colon cancers and in at least 30 of 36 colon cancer-derived cell lines. In colon-derived cell lines where Yap is overabundant, its depletion strongly reduces ß-catenin and Notch signaling and inhibits proliferation and survival. These findings demonstrate that Mst1 and Mst2 actively suppress Yap1 abundance and action in normal intestinal epithelium, an antiproliferative function that frequently is overcome in colon cancer through Yap1 polypeptide overabundance. The dispensability of Yap1 in normal intestinal homeostasis and its potent proliferative and prosurvival actions when overexpressed in colon cancer make it an attractive therapeutic target.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proliferación Celular , Colon/metabolismo , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Células Madre/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Western Blotting , Proteínas de Ciclo Celular , Línea Celular Tumoral , Colon/patología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Células HCT116 , Humanos , Inmunohistoquímica , Mucosa Intestinal/metabolismo , Intestinos/citología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Fosfoproteínas/genética , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Serina-Treonina Quinasa 3 , Células Madre/citología , Análisis de Matrices Tisulares , Factores de Transcripción , Proteínas Señalizadoras YAP
6.
Int J Gynecol Cancer ; 23(4): 615-21, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23552804

RESUMEN

OBJECTIVE: To evaluate pretherapy ribonucleotide reductase (RNR) expression and its effect on radiochemotherapeutic outcome in women with cervical cancer. METHODS/MATERIALS: Pretherapy RNR M1, M2, and M2b immunohistochemistry was done on cervical cancer specimens retrieved from women treated on Radiation Therapy Oncology Group (RTOG) 0116 and 0128 clinical trials. Enrollees of RTOG 0116 (node-positive stages IA-IVA) received weekly cisplatin (40 mg/m(2)) with amifostine (500 mg) and extended-field radiation then brachytherapy (85 Gy). Enrollees of RTOG 0128 (node-positive or bulky ≥5 cm, stages IB-IIA or stages IIB-IVA) received cisplatin (75 mg/m(2)) on days 1, 23, and 43 and 5-FU (1 g/m(2) for 4 days) during pelvic radiation then brachytherapy (85 Gy), plus celecoxib (400 mg twice daily, day 1 through 1 year). Disease-free survival (DFS) was estimated univariately by the Kaplan-Meier method. Cox proportional hazards models evaluated the impact of RNR immunoreactivity on DFS. RESULTS: Fifty-one tissue samples were analyzed: 13 from RTOG 0116 and 38 from RTOG 0128. M1, M2, and M2b overexpression (3+) frequencies were 2%, 80%, and 47%, respectively. Low-level (0-1+, n = 44/51) expression of the regulatory subunit M1 did not associate with DFS (P = 0.38). High (3+) M2 expression occurred in most (n = 41/51) but without impact alone on DFS (hazard ratio, 0.54; 95% confidence interval, 0.2-1.4; P = 0.20). After adjusting for M2b status, pelvic node-positive women had increased hazard for relapse or death (hazard ratio, 5.5; 95% confidence interval, 2.2-13.8; P = 0.0003). CONCLUSIONS: These results suggest that RNR subunit expression may discriminate cervical cancer phenotype and radiochemotherapy outcome. Future RNR biomarker studies are warranted.


Asunto(s)
Biomarcadores de Tumor/biosíntesis , Ribonucleótido Reductasas/biosíntesis , Neoplasias del Cuello Uterino/enzimología , Adulto , Anciano , Biomarcadores de Tumor/genética , Ensayos Clínicos Fase II como Asunto , Supervivencia sin Enfermedad , Femenino , Humanos , Persona de Mediana Edad , Fenotipo , Estudios Retrospectivos , Ribonucleótido Reductasas/genética , Investigación Biomédica Traslacional , Resultado del Tratamiento , Estados Unidos/epidemiología , Neoplasias del Cuello Uterino/genética , Neoplasias del Cuello Uterino/terapia
7.
Proc Natl Acad Sci U S A ; 107(6): 2592-7, 2010 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-20133777

RESUMEN

Protein tyrosine phosphatase receptor-type T (PTPRT) is the most frequently mutated tyrosine phosphatase in human cancers. However, the cell signaling pathways regulated by PTPRT largely remain to be elucidated. Here, we show that paxillin is a direct substrate of PTPRT and that PTPRT specifically regulates paxillin phosphorylation at tyrosine residue 88 (Y88) in colorectal cancer (CRC) cells. We engineered CRC cells homozygous for a paxillin Y88F knock-in mutant and found that these cells exhibit significantly reduced cell migration and impaired anchorage-independent growth, fail to form xenograft tumors in nude mice, and have decreased phosphorylation of p130CAS, SHP2, and AKT. PTPRT knockout mice that we generated exhibit increased levels of colonic paxillin phosphorylation at residue Y88 and are highly susceptible to carcinogen azoxymethane-induced colon tumor, providing critical in vivo evidence that PTPRT normally functions as a tumor suppressor. Moreover, similarly increased paxillin pY88 is also found as a common feature of human colon cancers. These studies reveal an important signaling pathway that plays a critical role in colorectal tumorigenesis.


Asunto(s)
Paxillin/metabolismo , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/metabolismo , Tirosina/metabolismo , Animales , Azoximetano , Western Blotting , Línea Celular , Línea Celular Tumoral , Proliferación Celular , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/patología , Femenino , Células HCT116 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Desnudos , Mutación , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/genética , Neoplasias Experimentales/metabolismo , Paxillin/genética , Paxillin/fisiología , Fosforilación , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Especificidad por Sustrato , Transfección , Trasplante Heterólogo , Tirosina/genética
8.
Gut ; 61(5): 665-72, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-21930730

RESUMEN

BACKGROUND: It is postulated that high serum levels of insulin and insulin growth factor 1 (IGF-1) mediate obesity-associated carcinogenesis. The relationship of insulin, IGF-1 and IGF binding proteins (IGFBP) with Barrett's oesophagus (BO) has not been well examined. METHODS: Serum levels of insulin and IGFBPs in patients with BO were compared with two separate control groups: subjects with gastro-oesophageal reflux disease (GORD) and screening colonoscopy controls. Fasting insulin, IGF-1 and IGFBPs were assayed in the serum of BO cases (n = 135), GORD (n = 135) and screening colonoscopy (n = 932) controls recruited prospectively at two academic hospitals. Logistic regression was used to estimate the risk of BO. RESULTS: Patients in the highest tertile of serum insulin levels had an increased risk of BO compared with colonoscopy controls (adjusted OR 2.02, 95% CI 1.15 to 3.54) but not compared with GORD controls (adjusted OR 1.55, 95% CI 0.76 to 3.15). Serum IGF-1 levels in the highest tertile were associated with an increased risk of BO (adjusted OR 4.05, 95% CI 2.01 to 8.17) compared with the screening colonoscopy control group but were not significantly different from the GORD control group (adjusted OR 0.57, 95% CI 0.27 to 1.17). IGFBP-1 levels in the highest tertile were inversely associated with a risk of BO in comparison with the screening colonoscopy controls (adjusted OR 0.11, 95% CI 0.05 to 0.24) but were not significantly different from the GORD control group (adjusted OR 1.04, 95% CI 0.49 to 2.16). IGFBP-3 levels in the highest tertile were inversely associated with the risk of BO compared with the GORD controls (OR 0.36, 95% CI 0.16 to 0.81) and also when compared with the colonoscopy controls (OR 0.40, 95% CI 0.20 to 0.79). CONCLUSIONS: These results provide support for the hypothesis that the insulin/IGF signalling pathways have a role in the development of BO.


Asunto(s)
Esófago de Barrett/sangre , Proteína 1 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Proteína 3 de Unión a Factor de Crecimiento Similar a la Insulina/sangre , Factor I del Crecimiento Similar a la Insulina/metabolismo , Insulina/sangre , Anciano , Esófago de Barrett/etiología , Estudios de Casos y Controles , Femenino , Reflujo Gastroesofágico/sangre , Humanos , Modelos Logísticos , Masculino , Persona de Mediana Edad , Análisis Multivariante , Obesidad/complicaciones , Estudios Prospectivos
9.
Int J Gynecol Cancer ; 22(9): 1463-9, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23051959

RESUMEN

OBJECTIVE: Ribonucleotide reductase (RNR) supplies deoxyribonucleotide diphosphates demanded by cells to repair radiation-induced DNA damage. Here, we investigate the impact of pretherapy RNR M1, M2, and M2b (p53R3) subunit level upon human cervical cancer radiochemosensitivity. MATERIALS/METHODS: Immunohistochemistry was performed on a tissue array comprised of 18 paired benign uterine cervix and stage IB2 cervical cancers to evaluate the relationship between cytosolic RNR M1, M2, and M2b staining intensity and radiochemotherapy cancer response. Patients underwent surgical hysterectomy (n = 8), or daily radiation (45 Gy), coadministered once-weekly cisplatin (40 mg/m), then low-dose rate brachytherapy (30 Gy) followed by adjuvant hysterectomy (n = 10). Radiochemotherapy response was determined by Response Evaluation Criteria In Solid Tumors version 1.0 criteria during brachytherapy. Cancer relapse rates and disease-free survival were calculated. RESULTS: M1, M2, and M2b antibody staining intensity was low (0-1+) in benign uterine cervical tissue. M1 and M2b immunoreactivity was 2+ or 3+ in most (13/18) cervical cancers. M2 immunoreactivity was 3+ in nearly all (16/18) cervical cancers. Cervical cancers overexpressing M1 and M2b had an increased hazard for incomplete radiochemotherapy response, relapse, and shortened disease-free survival. CONCLUSIONS: Ribonucleotide reductase subunit levels may predict human cervical cancer radiochemosensitivity and subsequent posttherapy cancer outcome. Further validation testing of RNR subunits as biomarkers for radiochemotherapy response is warranted.


Asunto(s)
Carcinoma de Células Escamosas/terapia , Quimioradioterapia , Tolerancia a Radiación , Ribonucleótido Reductasas/metabolismo , Neoplasias del Cuello Uterino/terapia , Adenocarcinoma/metabolismo , Adenocarcinoma/terapia , Adulto , Anciano , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/fisiología , Carcinoma de Células Escamosas/metabolismo , Femenino , Humanos , Histerectomía , Persona de Mediana Edad , Tolerancia a Radiación/fisiología , Ribonucleótido Reductasas/fisiología , Análisis de Matrices Tisulares , Insuficiencia del Tratamiento , Regulación hacia Arriba/fisiología , Neoplasias del Cuello Uterino/metabolismo
10.
Proc Natl Acad Sci U S A ; 106(23): 9409-13, 2009 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-19470469

RESUMEN

Pharmacologic inhibitors of the prostaglandin-synthesizing COX-2 oncogene prevent the development of premalignant human colon adenomas. However, resistance to treatment is common. In this study, we show that the adenoma prevention activity of the COX-2 inhibitor celecoxib requires the concomitant presence of the 15-hydroxyprostaglandin dehydrogenase (15-PGDH) tumor suppressor gene, and that loss of 15-PGDH expression imparts resistance to celecoxib's anti-tumor effects. We first demonstrate that the adenoma-preventive activity of celecoxib is abrogated in mice genetically lacking 15-PGDH. In FVB mice, celecoxib prevents 85% of azoxymethane-induced tumors >1 mm in size, but is essentially inactive in preventing tumor induction in 15-PGDH-null animals. Indeed, celecoxib treated 15-PGDH null animals develop more tumors than do celecoxib naive WT mice. In parallel with the loss of tumor prevention activity, celecoxib-mediated suppression of colonic PGE(2) levels is also markedly attenuated in 15-PGDH-null versus WT mice. Finally, as predicted by the murine models, humans with low colonic 15-PGDH levels also exhibit celecoxib resistance. Specifically, in a colon adenoma prevention trial, in all cases tested, individuals who developed new adenomas while receiving celecoxib treatment were also found as having low colonic 15-PGDH levels.


Asunto(s)
Adenoma/prevención & control , Neoplasias del Colon/prevención & control , Hidroxiprostaglandina Deshidrogenasas/genética , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Pirazoles/uso terapéutico , Sulfonamidas/uso terapéutico , Animales , Celecoxib , Colon/metabolismo , Colon/patología , Colonoscopía , Control de Medicamentos y Narcóticos , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/patología , Ratones , Ratones Noqueados , Pirazoles/metabolismo , Sulfonamidas/metabolismo
11.
Proc Natl Acad Sci U S A ; 106(31): 12921-5, 2009 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-19617566

RESUMEN

Aberrant glycosylation is a pathological alteration that is widespread in colon cancer, and usually accompanies the onset and progression of the disease. To date, the molecular mechanisms underlying aberrant glycosylation remain largely unknown. In this study, we identify somatic and germ-line mutations in the gene encoding for polypeptide N-acetylgalactosaminyltransferase 12 (GALNT12) in individuals with colon cancer. Biochemical analyses demonstrate that each of the 8 GALNT12 mutations identified inactivates the normal function of the GALNT enzyme in initiating mucin type O-linked protein glycosylation. Two of these inactivating GALNT12 mutations were identified as acquired somatic mutations in a set of 30 microsatellite stable colon tumors. Relative to background gene mutation rates, finding these somatic GALNT12 mutations was statistically significant at P < 0.001. Six additional inactivating GALNT12 mutations were detected as germ-line changes carried by patients with colon cancer; however, no inactivating variants were detected among cancer-free controls (P = 0.005). Notably, in 3 of the 6 individuals harboring inactivating germ-line GALNT12 mutations, both a colon cancer and a second independent epithelial cancer had developed. These findings suggest that genetic defects in the O-glycosylation pathway in part underlie aberrant glycosylation in colon cancers, and they contribute to the development of a subset of these malignancies.


Asunto(s)
Neoplasias del Colon/genética , Mutación de Línea Germinal , Mutación , N-Acetilgalactosaminiltransferasas/genética , Anciano , Animales , Línea Celular Tumoral , Glicosilación , Humanos , Ratones , Células 3T3 NIH
12.
PLoS One ; 17(5): e0268787, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35587945

RESUMEN

Emerging evidence implicates the eicosanoid molecule prostaglandin E2 (PGE2) in conferring a regenerative phenotype to multiple organ systems following tissue injury. As aging is in part characterized by loss of tissue stem cells' regenerative capacity, we tested the hypothesis that the prostaglandin-degrading enzyme 15-hydroxyprostaglandin dehydrogenase (15-PGDH) contributes to the diminished organ fitness of aged mice. Here we demonstrate that genetic loss of 15-PGDH (Hpgd) confers a protective effect on aging of murine hematopoietic and gastrointestinal (GI) tissues. Aged mice lacking 15-PGDH display increased hematopoietic output as assessed by peripheral blood cell counts, bone marrow and splenic stem cell compartments, and accelerated post-transplantation recovery compared to their WT counterparts. Loss of Hpgd expression also resulted in enhanced GI fitness and reduced local inflammation in response to colitis. Together these results suggest that 15-PGDH negatively regulates aged tissue regeneration, and that 15-PGDH inhibition may be a viable therapeutic strategy to ameliorate age-associated loss of organ fitness.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas , Envejecimiento/genética , Animales , Dinoprostona/metabolismo , Hidroxiprostaglandina Deshidrogenasas/genética , Ratones
13.
J Med Chem ; 65(22): 15327-15343, 2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36322935

RESUMEN

15-Prostaglandin dehydrogenase (15-PGDH) regulates the concentration of prostaglandin E2 in vivo. Inhibitors of 15-PGDH elevate PGE2 levels and promote tissue repair and regeneration. Here, we describe a novel class of quinoxaline amides that show potent inhibition of 15-PGDH, good oral bioavailability, and protective activity in mouse models of ulcerative colitis and recovery from bone marrow transplantation.


Asunto(s)
Hidroxiprostaglandina Deshidrogenasas , Quinoxalinas , Animales , Ratones , Colitis Ulcerosa/tratamiento farmacológico , Dinoprostona , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Quinoxalinas/farmacología
14.
Future Healthc J ; 9(3): 321-325, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36561817

RESUMEN

Introduction: The COVID-19 vaccination service is a key component in the UK approach to reducing disease morbidity and mortality. Groups within the population at increased risk of severe outcomes from COVID-19 overlap with groups that are less likely to take up the offer of vaccination. This article outlines some learning from approaches within a large vaccination centre in the UK to reduce inequalities. Solution: Continuous quality improvement processes were used to operationalise the mitigations to inequalities with vaccination uptake that were identified by a systematic equality impact assessment framework and continuous service feedback. Outcome: Quality improvement processes and community engagement enabled tailored mitigations to vaccination uptake. Engagement with community ambassadors strengthened community relationships and the co-creation of bespoke sessions encouraged vaccination uptake within specific groups. Conclusion: Recommendations for strengthening approaches to inequality reduction include having a systematic framework for assessment and mitigation of inequalities, embedding quality improvement, identifying resources, and taking a collaborative and co-design approach to services with underserved groups.

15.
Proc Natl Acad Sci U S A ; 105(42): 16224-9, 2008 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-18852474

RESUMEN

We have performed a genome-wide analysis of copy number changes in breast and colorectal tumors using approaches that can reliably detect homozygous deletions and amplifications. We found that the number of genes altered by major copy number changes, deletion of all copies or amplification to at least 12 copies per cell, averaged 17 per tumor. We have integrated these data with previous mutation analyses of the Reference Sequence genes in these same tumor types and have identified genes and cellular pathways affected by both copy number changes and point alterations. Pathways enriched for genetic alterations included those controlling cell adhesion, intracellular signaling, DNA topological change, and cell cycle control. These analyses provide an integrated view of copy number and sequencing alterations on a genome-wide scale and identify genes and pathways that could prove useful for cancer diagnosis and therapy.


Asunto(s)
Neoplasias de la Mama/genética , Neoplasias Colorrectales/genética , Amplificación de Genes/genética , Homocigoto , Eliminación de Gen , Transducción de Señal
16.
JCI Insight ; 6(6)2021 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-33600377

RESUMEN

The splenic microenvironment regulates hematopoietic stem and progenitor cell (HSPC) function, particularly during demand-adapted hematopoiesis; however, practical strategies to enhance splenic support of transplanted HSPCs have proved elusive. We have previously demonstrated that inhibiting 15-hydroxyprostaglandin dehydrogenase (15-PGDH), using the small molecule (+)SW033291 (PGDHi), increases BM prostaglandin E2 (PGE2) levels, expands HSPC numbers, and accelerates hematologic reconstitution after BM transplantation (BMT) in mice. Here we demonstrate that the splenic microenvironment, specifically 15-PGDH high-expressing macrophages, megakaryocytes (MKs), and mast cells (MCs), regulates steady-state hematopoiesis and potentiates recovery after BMT. Notably, PGDHi-induced neutrophil, platelet, and HSPC recovery were highly attenuated in splenectomized mice. PGDHi induced nonpathologic splenic extramedullary hematopoiesis at steady state, and pretransplant PGDHi enhanced the homing of transplanted cells to the spleen. 15-PGDH enzymatic activity localized specifically to macrophages, MK lineage cells, and MCs, identifying these cell types as likely coordinating the impact of PGDHi on splenic HSPCs. These findings suggest that 15-PGDH expression marks HSC niche cell types that regulate hematopoietic regeneration. Therefore, PGDHi provides a well-tolerated strategy to therapeutically target multiple HSC niches, promote hematopoietic regeneration, and improve clinical outcomes of BMT.


Asunto(s)
Células de la Médula Ósea/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hematopoyesis Extramedular/efectos de los fármacos , Hidroxiprostaglandina Deshidrogenasas/antagonistas & inhibidores , Regeneración , Bazo/efectos de los fármacos , Animales , Células de la Médula Ósea/citología , Femenino , Perfilación de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Bazo/enzimología , Bazo/metabolismo
17.
Cancer Epidemiol Biomarkers Prev ; 27(7): 728-736, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29769213

RESUMEN

Background: The Adenoma Prevention with Celecoxib (APC) Trial showed that cyclooxygenase-2 (Cox-2) inhibitor, celecoxib, decreased adenoma development in patients at high risk for colorectal cancer. A prospectively planned analysis of the APC Trial tested the hypothesis that expression of target enzymes in adenomas removed before beginning study treatment would identify individuals at high risk of adenoma development, and/or predict response to Cox-2 inhibition.Methods: Pre-treatment adenomas were examined using immunohistochemistry to assess expression of Cox-2 (high vs. low) and 15-prostaglandin dehydrogenase (15-PGDH, presence vs. loss). The Mantel-Cox test evaluated whether these markers predicted benefit from celecoxib for reduction of adenoma detection.Results: Patients whose pre-treatment adenomas demonstrated elevated Cox-2 achieved the greatest adenoma reduction with celecoxib treatment [RR, 0.37; 95% confidence interval (CI), 0.22-0.61; P = 0.0001]. This reduction was less in the low Cox-2 category (RR, 0.64; 95% CI, 0.56-0.73). Patients whose pre-treatment adenomas showed 15-PGDH loss had a similar treatment-associated reduction in adenoma detection (RR, 0.60; 95% CI, 0.52-0.69; P < 0.0001). In contrast, patients with intact tumor 15-PGDH expression did not significantly benefit from celecoxib (RR, 0.73; 95% CI, 0.47-1.12; P = 0.15). However, subset analysis suggested that this lack of response to celecoxib was confined to those patients with 15-PGDH intact tumors who were also using cardioprotective aspirin.Conclusions: The expression of Cox-2 and 15-PGDH in pre-treatment adenomas provides predictive information in patients treated with celecoxib for prevention of colorectal adenomas.Impact: The results of this study show that Cox-2 and 15-PGDH are characteristics of colorectal adenomas that may be used to predict nonsteroidal anti-inflammatory drug chemoprevention efficacy. Cancer Epidemiol Biomarkers Prev; 27(7); 728-36. ©2018 AACR.


Asunto(s)
Adenoma/metabolismo , Celecoxib/uso terapéutico , Quimioprevención/métodos , Ciclooxigenasa 2/metabolismo , Hidroxiprostaglandina Deshidrogenasas/metabolismo , Celecoxib/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Estudios Prospectivos
19.
Cancer Res ; 63(7): 1568-75, 2003 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-12670906

RESUMEN

To identify potential effectors of transforming growth factor (TGF)-beta-mediated suppression of colon cancer, we used GeneChip expression microarrays to identify TGF-beta-induced genes in VACO 330, a nontransformed TGF-beta-sensitive cell line derived from a human adenomatous colon polyp. PMEPA1 was identified as a gene highly up-regulated by TGF-beta treatment of VACO 330. Northern blot analysis confirmed TGF-beta induction of PMEPA1 in VACO 330, as well as a panel of three other TGF-beta-sensitive colon cell lines. PMEPA1 induction could be detected as early as 2 h after TGF-beta treatment and was not inhibited by pretreatment of cells with cycloheximide, suggesting that PMEPA1 is a direct target of TGF-beta signaling. Wild-type PMEPA1 and an alternative splice variant lacking the putative transmembrane domain were encoded by the PMEPA1 locus and were shown by epitope tagging to encode proteins with differing subcellular localization. Both variants were found to be expressed in normal colonic epithelium, and both were shown to be induced by TGF-beta. Consistent with TGF-beta playing a role in terminal differentiation of colonocytes, in situ hybridization of normal colonic epithelium localized PMEPA1 expression to nonproliferating, terminally differentiated epithelium located at the top of colonic crypts. Intriguingly, in situ hybridization and Northern blot analysis showed that the expression of PMEPA1 was well maintained both in colon cancer primary tumors and in colon cancer liver metastases. PMEPA1 is thus a novel TGF-beta-induced marker of a differentiated crypt cell population. Moreover, as PMEPA1 expression is maintained, presumptively in a TGF-beta-independent manner after malignant transformation and metastasis, it demonstrates that even late colon cancers retain a strong capacity to execute many steps of the normal colonic differentiation program.


Asunto(s)
Neoplasias del Colon/metabolismo , Proteínas de la Membrana/biosíntesis , Factor de Crecimiento Transformador beta/fisiología , Empalme Alternativo , Secuencia de Aminoácidos , Secuencia de Bases , Diferenciación Celular/genética , División Celular/fisiología , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Colon/citología , Colon/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundario , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Datos de Secuencia Molecular , Isoformas de Proteínas , Transducción de Señal/fisiología , Fracciones Subcelulares/metabolismo , Transfección , Factor de Crecimiento Transformador beta/farmacología , Regulación hacia Arriba
20.
Oncogene ; 21(1): 78-86, 2002 Jan 03.
Artículo en Inglés | MEDLINE | ID: mdl-11791178

RESUMEN

The aim of this study was to determine whether constitutive ErbB2 activation controls growth and apoptosis in colon cancer cells. Growth arrested GEO cells showed constitutive activation of ErbB2 in the absence of exogenous growth factors or serum supplementation. Higher levels of heregulin and ErbB2 activation were observed in the growth-arrested state and cell cycle re-entry was independent of exogenous growth factors. Blockade of ErbB2 activation by heregulin neutralizing antibodies and by AG879 resulted in prevention of cell cycle re-entry. This indicated that autocrine heregulin activity was responsible for growth factor independence and for cell cycle re-entry. Activation of ErbB2 was the result of heregulin mediated interaction with ErbB3 and generated downstream activation of the ERK and the PI3K/AKT pathways. Heregulin neutralizing antibody treatment of growth arrested GEO cells also generated apoptosis as reflected by PARP cleavage and DNA fragmentation indicating a cell survival signal was also induced by the constitutively activated ErbB2. The activation of AKT but not the MAPK pathway was responsible for cell survival in these cells.


Asunto(s)
Adenocarcinoma/patología , Apoptosis/fisiología , Comunicación Autocrina , Neoplasias del Colon/patología , Proteínas de Neoplasias/fisiología , Neurregulina-1/fisiología , Transducción de Señal/fisiología , Ciclo Celular/efectos de los fármacos , Cromonas/farmacología , Medios de Cultivo/farmacología , Medio de Cultivo Libre de Suero , Fragmentación del ADN , Dimerización , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Genes erbB-2 , Sustancias de Crecimiento/farmacología , Sistema de Señalización de MAP Quinasas , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Morfolinas/farmacología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/inmunología , Neurregulina-1/genética , Neurregulina-1/inmunología , Pruebas de Neutralización , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/fisiología , Proteínas Proto-Oncogénicas c-akt , Receptor ErbB-3/fisiología , Transducción de Señal/efectos de los fármacos , Sirolimus/farmacología , Transfección , Factor de Crecimiento Transformador alfa/biosíntesis , Factor de Crecimiento Transformador alfa/genética , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA