Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
NPJ Breast Cancer ; 8(1): 84, 2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35853889

RESUMEN

There is growing evidence that germline mutations in certain genes influence cancer susceptibility, tumor evolution, as well as clinical outcomes. Identification of a disease-causing genetic variant enables testing and diagnosis of at-risk individuals. For breast cancer, several genes such as BRCA1, BRCA2, PALB2, ATM, and CHEK2 act as high- to moderate-penetrance cancer susceptibility genes. Genotyping of these genes informs genetic risk assessment and counseling, as well as treatment and management decisions in the case of high-penetrance genes. TGFBR1*6A (rs11466445) is a common variant of the TGF-ß receptor type I (TGFBR1) that has a global minor allelic frequency (MAF) of 0.051 according to the 1000 Genomes Project Consortium. It is emerging as a high frequency, low penetrance tumor susceptibility allele associated with increased cancer risk among several cancer types. The TGFBR1*6A allele has been associated with increased breast cancer risk in women, OR 1.15 (95% CI 1.01-1.31). Functionally, TGFBR1*6A promotes breast cancer cell proliferation, migration, and invasion through the regulation of the ERK pathway and Rho-GTP activation. This review discusses current findings on the genetic, functional, and mechanistic associations between TGFBR1*6A and breast cancer risk and proposes future directions as it relates to genetic association studies and mechanisms of action for tumor growth, metastasis, and immune suppression.

2.
EBioMedicine ; 44: 209-224, 2019 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-31160272

RESUMEN

BACKGROUND: Administration of amplitude modulated 27·12 MHz radiofrequency electromagnetic fields (AM RF EMF) by means of a spoon-shaped applicator placed on the patient's tongue is a newly approved treatment for advanced hepatocellular carcinoma (HCC). The mechanism of action of tumour-specific AM RF EMF is largely unknown. METHODS: Whole body and organ-specific human dosimetry analyses were performed. Mice carrying human HCC xenografts were exposed to AM RF EMF using a small animal AM RF EMF exposure system replicating human dosimetry and exposure time. We performed histological analysis of tumours following exposure to AM RF EMF. Using an agnostic genomic approach, we characterized the mechanism of action of AM RF EMF. FINDINGS: Intrabuccal administration results in systemic delivery of athermal AM RF EMF from head to toe at levels lower than those generated by cell phones held close to the body. Tumour shrinkage results from differentiation of HCC cells into quiescent cells with spindle morphology. AM RF EMF targeted antiproliferative effects and cancer stem cell inhibiting effects are mediated by Ca2+ influx through Cav3·2 T-type voltage-gated calcium channels (CACNA1H) resulting in increased intracellular calcium concentration within HCC cells only. INTERPRETATION: Intrabuccally-administered AM RF EMF is a systemic therapy that selectively block the growth of HCC cells. AM RF EMF pronounced inhibitory effects on cancer stem cells may explain the exceptionally long responses observed in several patients with advanced HCC. FUND: Research reported in this publication was supported by the National Cancer Institute's Cancer Centre Support Grant award number P30CA012197 issued to the Wake Forest Baptist Comprehensive Cancer Centre (BP) and by funds from the Charles L. Spurr Professorship Fund (BP). DWG is supported by R01 AA016852 and P50 AA026117.


Asunto(s)
Canales de Calcio Tipo T/metabolismo , Calcio/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/terapia , Magnetoterapia , Animales , Bloqueadores de los Canales de Calcio/farmacología , Carcinoma Hepatocelular/patología , Modelos Animales de Enfermedad , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/patología , Magnetoterapia/métodos , Ratones , Células Madre Neoplásicas/metabolismo , Especificidad de Órganos , ARN Interferente Pequeño/genética , Radiometría , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Trans Am Clin Climatol Assoc ; 125: 300-12, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25125747

RESUMEN

Transforming growth factor beta (TGF-ß) is a potent inhibitor of cell growth. TGFBR1 6A is a polymorphism consisting of a 9-base pair in-frame deletion within exon 1 of the type I TGF-ß receptor (TGFBR1), which results in a receptor with decreased TGF-ß signaling capability. The discovery of an association between TGFBR1*6A and cancer susceptibility led to the hypothesis that hypomorphic variants of the TGF-ß signaling pathway may predispose to the development of cancer. This hypothesis was tested in vivo with the development of a mouse model of Tgfbr1 haploinsufficiency. Tgfbr1 (+/-) mice developed twice as many intestinal tumors as Tgfbr1 (+/+). Tgfbr1 haploinsufficiency was also associated with early onset adenocarcinoma and increased tumor cell proliferation. A case control study identified two haplotypes associated with constitutively decreased TGFBR1 and substantially increased colorectal cancer risk indicating that TGFBR1 may act as a potent modifier of cancer risk.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Transformación Celular Neoplásica/metabolismo , Neoplasias/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento Transformadores beta/metabolismo , Animales , Biomarcadores de Tumor/deficiencia , Biomarcadores de Tumor/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Predisposición Genética a la Enfermedad , Humanos , Ratones Noqueados , Neoplasias/genética , Neoplasias/patología , Fenotipo , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta , Receptores de Factores de Crecimiento Transformadores beta/deficiencia , Receptores de Factores de Crecimiento Transformadores beta/genética , Factores de Riesgo , Transducción de Señal , Factor de Crecimiento Transformador beta/metabolismo
5.
Chin J Cancer ; 32(11): 573-81, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24206915

RESUMEN

In the past century, there have been many attempts to treat cancer with low levels of electric and magnetic fields. We have developed noninvasive biofeedback examination devices and techniques and discovered that patients with the same tumor type exhibit biofeedback responses to the same, precise frequencies. Intrabuccal administration of 27.12 MHz radiofrequency (RF) electromagnetic fields (EMF), which are amplitude-modulated at tumor-specific frequencies, results in long-term objective responses in patients with cancer and is not associated with any significant adverse effects. Intrabuccal administration allows for therapeutic delivery of very low and safe levels of EMF throughout the body as exemplified by responses observed in the femur, liver, adrenal glands, and lungs. In vitro studies have demonstrated that tumor-specific frequencies identified in patients with various forms of cancer are capable of blocking the growth of tumor cells in a tissue- and tumor-specific fashion. Current experimental evidence suggests that tumor-specific modulation frequencies regulate the expression of genes involved in migration and invasion and disrupt the mitotic spindle. This novel targeted treatment approach is emerging as an appealing therapeutic option for patients with advanced cancer given its excellent tolerability. Dissection of the molecular mechanisms accounting for the anti-cancer effects of tumor-specific modulation frequencies is likely to lead to the discovery of novel pathways in cancer.


Asunto(s)
Campos Electromagnéticos , Magnetoterapia , Neoplasias/terapia , Carcinoma Hepatocelular/terapia , Proliferación Celular/efectos de la radiación , Humanos , Neoplasias Hepáticas/terapia , Magnetoterapia/efectos adversos , Neoplasias/diagnóstico , Neoplasias/patología , Dosis de Radiación , Ondas de Radio , Neoplasias de la Tiroides/terapia , Resultado del Tratamiento
6.
Cancer Genet ; 205(1-2): 25-33, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22429595

RESUMEN

Recent genome-wide association studies have identified multiple regions at 8q24 that confer susceptibility to many cancers. In our previous work, we showed that the colorectal cancer (CRC) risk variant rs6983267 at 8q24 resides within a TCF4 binding site at the MYC-335 enhancer, with the risk allele G having a stronger binding capacity and Wnt responsiveness. Here, we searched for other potential functional variants within MYC-335. Genetic variation within MYC-335 was determined in samples from individuals of European, African, and Asian descent, with emphasis on variants in putative transcription factor binding sites. A 2-bp GA deletion rs67491583 was found to affect a growth factor independent (GFI) binding site and was present only in individuals with African ancestry. Chromatin immunoprecipitation performed in heterozygous cells showed that the GA deletion had an ability to reduce binding of the transcriptional repressors GFI1 and GFI1b. Screening of 1,027 African American colorectal cancer cases and 1,773 healthy controls did not reveal evidence for association (odds ratio: 1.17, 95% confidence interval: 0.97-1.41, P = 0.095). In this study, rs67491583 was identified as another functional variant in the CRC-associated enhancer MYC-335, but further studies are needed to establish the role of rs67491583 in the colorectal cancer predisposition of African Americans.


Asunto(s)
Carcinoma/genética , Cromosomas Humanos Par 8 , Neoplasias Colorrectales/genética , Genes myc/genética , Polimorfismo de Nucleótido Simple/fisiología , Adulto , Negro o Afroamericano/genética , Negro o Afroamericano/estadística & datos numéricos , Pueblo Asiatico/genética , Pueblo Asiatico/estadística & datos numéricos , Carcinoma/etnología , Estudios de Casos y Controles , Cromosomas Humanos Par 8/genética , Estudios de Cohortes , Neoplasias Colorrectales/etnología , Elementos de Facilitación Genéticos/genética , Elementos de Facilitación Genéticos/fisiología , Femenino , Regulación Neoplásica de la Expresión Génica , Frecuencia de los Genes , Genes myc/fisiología , Genotipo , Células HEK293 , Células HeLa , Humanos , Transfección , Células Tumorales Cultivadas , Población Blanca/genética , Población Blanca/estadística & datos numéricos
7.
J Exp Clin Cancer Res ; 29: 57, 2010 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-20500843

RESUMEN

PURPOSE: Constitutively decreased TGFBR1 allelic expression is emerging as a potent modifier of colorectal cancer risk in mice and humans. This phenotype was first observed in mice, then in lymphoblastoid cell lines from patients with microsatellite stable colorectal tumors. PATIENTS AND METHODS: We assessed the frequency of constitutively decreased TGFBR1 allelic expression and association with SNPs covering the TGFBR1 locus using RNA and DNA extracted from the peripheral blood lymphocytes of 118 consecutive patients with biopsy-proven adenocarcinoma of the colon or the rectum. RESULTS: We found that 11(9.3%) of 118 patients exhibited decreased TGFBR1 allelic expression (TGFBR1 ASE). TGFBR1 ASE was strongly associated with three SNPs in linkage disequilibrium with each other: rs7034462 (p = 7.2 x 10-4), TGFBR1*6A (p = 1.6 x 10-4) and rs11568785 (p = 1.4 x 10-4). CONCLUSION: These results confirm the high prevalence of constitutively decreased TGFBR1 allelic expression among patients with colorectal cancer. The association of this phenotype with TGFBR1*6A, rs7034462 and rs1156875 suggests an association between TGFBR1 SNPs and colorectal cancer, which warrants additional studies.


Asunto(s)
Adenocarcinoma/genética , Neoplasias Colorrectales/genética , Polimorfismo de Nucleótido Simple/genética , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Adenocarcinoma/patología , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Western Blotting , Neoplasias Colorrectales/patología , Estudios Transversales , ADN de Neoplasias/genética , Femenino , Haplotipos/genética , Humanos , Masculino , Persona de Mediana Edad , Reacción en Cadena de la Polimerasa , Pronóstico , ARN Mensajero/genética , ARN Neoplásico/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta
8.
Science ; 321(5894): 1361-5, 2008 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-18703712

RESUMEN

Much of the genetic predisposition to colorectal cancer (CRC) in humans is unexplained. Studying a Caucasian-dominated population in the United States, we showed that germline allele-specific expression (ASE) of the gene encoding transforming growth factor-beta (TGF-beta) type I receptor, TGFBR1, is a quantitative trait that occurs in 10 to 20% of CRC patients and 1 to 3% of controls. ASE results in reduced expression of the gene, is dominantly inherited, segregates in families, and occurs in sporadic CRC cases. Although subtle, the reduction in constitutive TGFBR1 expression alters SMAD-mediated TGF-beta signaling. Two major TGFBR1 haplotypes are predominant among ASE cases, which suggests ancestral mutations, but causative germline changes have not been identified. Conservative estimates suggest that ASE confers a substantially increased risk of CRC (odds ratio, 8.7; 95% confidence interval, 2.6 to 29.1), but these estimates require confirmation and will probably show ethnic differences.


Asunto(s)
Neoplasias Colorrectales/genética , Expresión Génica , Predisposición Genética a la Enfermedad , Proteínas Serina-Treonina Quinasas/genética , Receptores de Factores de Crecimiento Transformadores beta/genética , Regiones no Traducidas 3' , Adulto , Anciano , Anciano de 80 o más Años , Alelos , Línea Celular , Femenino , Haplotipos , Heterocigoto , Humanos , Desequilibrio de Ligamiento , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Carácter Cuantitativo Heredable , Receptor Tipo I de Factor de Crecimiento Transformador beta , Factores de Riesgo , Transducción de Señal , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...