Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
1.
NPJ Breast Cancer ; 8(1): 84, 2022 Jul 19.
Article in English | MEDLINE | ID: mdl-35853889

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-31160272

ABSTRACT

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.


Subject(s)
Calcium Channels, T-Type/metabolism , Calcium/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Magnetic Field Therapy , Animals , Calcium Channel Blockers/pharmacology , Carcinoma, Hepatocellular/pathology , Disease Models, Animal , Gene Knockdown Techniques , Humans , Liver Neoplasms/pathology , Magnetic Field Therapy/methods , Mice , Neoplastic Stem Cells/metabolism , Organ Specificity , RNA, Small Interfering/genetics , Radiometry , Treatment Outcome , Xenograft Model Antitumor Assays
4.
Trans Am Clin Climatol Assoc ; 125: 300-12, 2014.
Article in English | MEDLINE | ID: mdl-25125747

ABSTRACT

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.


Subject(s)
Biomarkers, Tumor/metabolism , Cell Transformation, Neoplastic/metabolism , Neoplasms/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Biomarkers, Tumor/deficiency , Biomarkers, Tumor/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/pathology , Genetic Predisposition to Disease , Humans , Mice, Knockout , Neoplasms/genetics , Neoplasms/pathology , Phenotype , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type I , Receptors, Transforming Growth Factor beta/deficiency , Receptors, Transforming Growth Factor beta/genetics , Risk Factors , Signal Transduction , Transforming Growth Factor beta/metabolism
5.
Chin J Cancer ; 32(11): 573-81, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24206915

ABSTRACT

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.


Subject(s)
Electromagnetic Fields , Magnetic Field Therapy , Neoplasms/therapy , Carcinoma, Hepatocellular/therapy , Cell Proliferation/radiation effects , Humans , Liver Neoplasms/therapy , Magnetic Field Therapy/adverse effects , Neoplasms/diagnosis , Neoplasms/pathology , Radiation Dosage , Radio Waves , Thyroid Neoplasms/therapy , Treatment Outcome
6.
Cancer Genet ; 205(1-2): 25-33, 2012.
Article in English | MEDLINE | ID: mdl-22429595

ABSTRACT

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.


Subject(s)
Carcinoma/genetics , Chromosomes, Human, Pair 8 , Colorectal Neoplasms/genetics , Genes, myc/genetics , Polymorphism, Single Nucleotide/physiology , Adult , Black or African American/genetics , Black or African American/statistics & numerical data , Asian People/genetics , Asian People/statistics & numerical data , Carcinoma/ethnology , Case-Control Studies , Chromosomes, Human, Pair 8/genetics , Cohort Studies , Colorectal Neoplasms/ethnology , Enhancer Elements, Genetic/genetics , Enhancer Elements, Genetic/physiology , Female , Gene Expression Regulation, Neoplastic , Gene Frequency , Genes, myc/physiology , Genotype , HEK293 Cells , HeLa Cells , Humans , Transfection , Tumor Cells, Cultured , White People/genetics , White People/statistics & numerical data
7.
J Exp Clin Cancer Res ; 29: 57, 2010 May 25.
Article in English | MEDLINE | ID: mdl-20500843

ABSTRACT

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.


Subject(s)
Adenocarcinoma/genetics , Colorectal Neoplasms/genetics , Polymorphism, Single Nucleotide/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Alleles , Blotting, Western , Colorectal Neoplasms/pathology , Cross-Sectional Studies , DNA, Neoplasm/genetics , Female , Haplotypes/genetics , Humans , Male , Middle Aged , Polymerase Chain Reaction , Prognosis , RNA, Messenger/genetics , RNA, Neoplasm/genetics , Receptor, Transforming Growth Factor-beta Type I
8.
Science ; 321(5894): 1361-5, 2008 Sep 05.
Article in English | MEDLINE | ID: mdl-18703712

ABSTRACT

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.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression , Genetic Predisposition to Disease , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , 3' Untranslated Regions , Adult , Aged , Aged, 80 and over , Alleles , Cell Line , Female , Haplotypes , Heterozygote , Humans , Linkage Disequilibrium , Male , Middle Aged , Polymorphism, Single Nucleotide , Quantitative Trait, Heritable , Receptor, Transforming Growth Factor-beta Type I , Risk Factors , Signal Transduction , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL