1
|
Abstract
The kexin-like proprotein convertases perform the initial proteolytic cleavages that ultimately generate a variety of different mature peptide and proteins, ranging from brain neuropeptides to endocrine peptide hormones, to structural proteins, among others. In this review, we present a general introduction to proprotein convertase structure and biochemistry, followed by a comprehensive discussion of each member of the kexin-like subfamily of proprotein convertases. We summarize current knowledge of human proprotein convertase insufficiency syndromes, including genome-wide analyses of convertase polymorphisms, and compare these to convertase null and mutant mouse models. These mouse models have illuminated our understanding of the roles specific convertases play in human disease and have led to the identification of convertase-specific substrates; for example, the identification of procorin as a specific PACE4 substrate in the heart. We also discuss the limitations of mouse null models in interpreting human disease, such as differential precursor cleavage due to species-specific sequence differences, and the challenges presented by functional redundancy among convertases in attempting to assign specific cleavages and/or physiological roles. However, in most cases, knockout mouse models have added substantively both to our knowledge of diseases caused by human proprotein convertase insufficiency and to our appreciation of their normal physiological roles, as clearly seen in the case of the furin, proprotein convertase 1/3, and proprotein convertase 5/6 mouse models. The creation of more sophisticated mouse models with tissue- or temporally-restricted expression of specific convertases will improve our understanding of human proprotein convertase insufficiency and potentially provide support for the emerging concept of therapeutic inhibition of convertases.
Collapse
Affiliation(s)
- Manita Shakya
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
2
|
Xicola RM, Bontu S, Doyle BJ, Rawson J, Garre P, Lee E, de la Hoya M, Bessa X, Clofent J, Bujanda L, Balaguer F, Castellví-Bel S, Alenda C, Jover R, Ruiz-Ponte C, Syngal S, Andreu M, Carracedo A, Castells A, Newcomb PA, Lindor N, Potter JD, Baron JA, Ellis NA, Caldes T, LLor X. Association of a let-7 miRNA binding region of TGFBR1 with hereditary mismatch repair proficient colorectal cancer (MSS HNPCC). Carcinogenesis 2016; 37:751-758. [PMID: 27234654 PMCID: PMC4967215 DOI: 10.1093/carcin/bgw064] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 04/24/2016] [Accepted: 05/15/2016] [Indexed: 12/30/2022] Open
Abstract
The purpose of this study was to identify novel colorectal cancer (CRC)-causing alleles in unexplained familial CRC cases. In order to do so, coding regions in five candidate genes (MGMT, AXIN2, CTNNB1, TGFBR1 and TGFBR2) were sequenced in 11 unrelated microsatellite-stable hereditary non-polyposis CRC (MSS HNPCC) cases. Selected genetic variants were genotyped in a discovery set of 27 MSS HNPCC cases and 85 controls. One genetic variant, rs67687202, in TGFBR1 emerged as significant (P = 0.002), and it was genotyped in a replication set of 87 additional MSS HNPCC-like cases and 338 controls where it was also significantly associated with MSS HNPCC cases (P = 0.041). In the combined genotype data, rs67687202 was associated with a moderate increase in CRC risk (OR = 1.68; 95% CI = 1.13-2.50; P = 0.010). We tested a highly correlated SNP rs868 in 723 non-familial CRC cases compared with 629 controls, and it was not significantly associated with CRC risk (P = 0.370). rs868 is contained in a let-7 miRNA binding site in the 3'UTR of TGFBR1, which might provide a functional basis for the association in MSS HNPCC. In luciferase assays, the risk-associated allele for rs868 was associated with half the luciferase expression in the presence of miRNA let-7b-5p compared with protective allele, suggesting more binding of let-7b-5p and less TGFBR1 expression. Thus, rs868 potentially is a CRC risk-causing allele. Our results support the concept that rs868 is associated with lower TGFBR1 expression thereby increasing CRC risk.
Collapse
Affiliation(s)
- Rosa M Xicola
- Department of Medicine and Cancer Center, Yale University, New Haven, CT, USA
| | - Sneha Bontu
- Department of Medicine and Cancer Center, Yale University, New Haven, CT, USA
| | - Brian J Doyle
- Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Jamie Rawson
- Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Pilar Garre
- Laboratorio de Oncología Molecular, Hospital Clinico San Carlos, IdISSC , Madrid, Spain
| | - Esther Lee
- Department of Medicine and Cancer Center, University of Illinois at Chicago, Chicago, IL, USA
| | - Miguel de la Hoya
- Laboratorio de Oncología Molecular, Hospital Clinico San Carlos, IdISSC , Madrid, Spain
| | - Xavier Bessa
- Department of Gastroenterology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Joan Clofent
- Department of Gastroenterology, Hospital de Sagunto, Sagunto, Valencia, Spain
| | - Luis Bujanda
- Department of Gastroenterology, Hospital Donostia/Instituto Biodonostia, CIBERehd, Universidad del País Vasco (UPV/EHU), San Sebastian, Spain
| | - Francesc Balaguer
- Department of Gastroenterology, Hospital Clinic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - Sergi Castellví-Bel
- Department of Gastroenterology, Hospital Clinic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | - Cristina Alenda
- Department of Gastroenterology and Department of Pathology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Rodrigo Jover
- Department of Gastroenterology and Department of Pathology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Clara Ruiz-Ponte
- Fundación Pública Galega de Medicina Xenómica (FPGMX)-SERGAS, Grupo de Medicina Xenómica, IDIS, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Santiago de Compostela, Galiza, Spain
| | - Sapna Syngal
- Division of Population Sciences, Dana-Farber Cancer Institute, Boston, MA, USA Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Montserrat Andreu
- Department of Gastroenterology, Hospital del Mar, Barcelona, Catalonia, Spain
| | - Angel Carracedo
- Fundación Pública Galega de Medicina Xenómica (FPGMX)-SERGAS, Grupo de Medicina Xenómica, IDIS, Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERer), Santiago de Compostela, Galiza, Spain
| | - Antoni Castells
- Department of Gastroenterology, Hospital Clinic, IDIBAPS, CIBERehd, University of Barcelona, Barcelona, Spain
| | | | - Noralane Lindor
- Department of Health Sciences Research, Mayo Clinic, Scottsdale, AZ, USA
| | - John D Potter
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA Department of Epidemiology, University of Washington, Seattle, WA, USA Centre for Public Health Research, Wellington, New Zealand
| | - John A Baron
- Department of Biostatistics and Epidemiology, Dartmouth College, Lebanon, NH, USA
| | - Nathan A Ellis
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Trinidad Caldes
- Laboratorio de Oncología Molecular, Hospital Clinico San Carlos, IdISSC , Madrid, Spain
| | - Xavier LLor
- Department of Medicine and Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
3
|
Lamar KM, Miller T, Dellefave-Castillo L, McNally EM. Genotype-Specific Interaction of Latent TGFβ Binding Protein 4 with TGFβ. PLoS One 2016; 11:e0150358. [PMID: 26918958 PMCID: PMC4769137 DOI: 10.1371/journal.pone.0150358] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 02/12/2016] [Indexed: 01/06/2023] Open
Abstract
Latent TGFβ binding proteins are extracellular matrix proteins that bind latent TGFβ to form the large latent complex. Nonsynonymous polymorphisms in LTBP4, a member of the latent TGFβ binding protein gene family, have been linked to several human diseases, underscoring the importance of TGFβ regulation for a range of phenotypes. Because of strong linkage disequilibrium across the LTBP4 gene, humans have two main LTBP4 alleles that differ at four amino acid positions, referred to as IAAM and VTTT for the encoded residues. VTTT is considered the “risk” allele and associates with increased intracellular TGFβ signaling and more deleterious phenotypes in muscular dystrophy and other diseases. We now evaluated LTBP4 nsSNPs in dilated cardiomyopathy, a distinct disorder associated with TGFβ signaling. We stratified based on self-identified ethnicity and found that the LTBP4 VTTT allele is associated with increased risk of dilated cardiomyopathy in European Americans extending the diseases that associate with LTBP4 genotype. However, the association of LTBP4 SNPs with dilated cardiomyopathy was not observed in African Americans. To elucidate the mechanism by which LTBP4 genotype exerts this differential effect, TGFβ’s association with LTBP4 protein was examined. LTBP4 protein with the IAAM residues bound more latent TGFβ compared to the LTBP4 VTTT protein. Together these data provide support that LTBP4 genotype exerts its effect through differential avidity for TGFβ accounting for the differences in TGFβ signaling attributed to these two alleles.
Collapse
Affiliation(s)
- Kay-Marie Lamar
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Tamari Miller
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
| | - Lisa Dellefave-Castillo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Elizabeth M. McNally
- Department of Human Genetics, The University of Chicago, Chicago, Illinois, United States of America
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
4
|
Ibrahim T, Yazbeck C, Maalouly G, Baz M, Haddad F, Sabbagh C, Chahine G. TGFBR1*6A polymorphism in sporadic and familial colorectal Carcinoma: a case-control study and systematic literature review. J Gastrointest Cancer 2015; 45:441-7. [PMID: 24880985 DOI: 10.1007/s12029-014-9625-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND The role of genetic factors in colorectal cancer pathogenesis is widely accepted. Polymorphisms are actually thought to play a role in the unexplained colorectal cancer (CRC) susceptibility. There is conflicting data regarding the role of the transforming growth factor beta receptor 1 polymorphism 6A (TGFBR1*6A) in the increased incidence of CRC. PURPOSE Our aim is to test the association between this polymorphism and sporadic/familial CRC in the Lebanese population paying attention to lead time bias in the control group. This is a case-control study conducted in two Lebanese hospital centers. MATERIALS AND METHODS Cases were diagnosed with CRC during the period of 1 year prior to the study. Controls were healthy subjects aged >50 years with a history of normal colonoscopy during the period of 5 years prior to the beginning of the study. A total of 96 cases (57 sporadic/39 familial) and 97 controls were genotyped. The odds ratios for 6A carrier status was statistically significant for sporadic CRC, odds ratio (OR) = 2.314 (95 % confidence interval (CI) 1.030-5.195) but not for familial CRC. RESULTS No association was found between 6A carrier status and mean age at diagnosis of CRC. This is the first article in the literature to evaluate the association between 6A polymorphism and total, sporadic, and familial CRC in a single study with reduction of bias in the control group. Results are in conjunction with other studies and meta-analysis.
Collapse
Affiliation(s)
- Tony Ibrahim
- Hemato-Oncology Department, Hotel Dieu de France teaching Hospital of Saint Joseph University, 11-5076, Riad El Solh-Beirut, 1107 2180, Beirut, Lebanon,
| | | | | | | | | | | | | |
Collapse
|
5
|
Association between Int7G24A rs334354 polymorphism and cancer risk: a meta-analysis of case-control studies. Sci Rep 2015; 5:11350. [PMID: 26074400 PMCID: PMC4466893 DOI: 10.1038/srep11350] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 05/22/2015] [Indexed: 11/27/2022] Open
Abstract
Accumulating evidences have suggested the potential association between Int7G24A
(rs334354) polymorphism and cancer risk. However, results from epidemiological
studies are controversial. We thus conducted this meta-analysis to clarify the
association. Relevant studies were identified on electronic databases according to
the inclusion criteria. A total of 13 case-control studies containing 4092 cases and
5909 controls were included in our meta-analysis. Odds ratios (ORs) with 95%
confidence intervals (CIs) were applied to assess the association. The results of
the overall population had suggested that Int7G24A polymorphism had an increased
risk for cancer, reaching significant levels in the 2 genetic models (allele model,
OR = 1.25, 95% CI 1.09-1.42,
P = 0.001; dominant model,
OR = 1.24, 95% CI 1.06-1.46,
P < 0.008). Besides, significant association
was found among Asian population (allele model, OR = 1.27,
95% CI 1.11-1.45, P < 0.001; dominant model,
OR = 1.28, 95% CI 1.11-1.49,
P < 0.001), whereas there was non-significant
relationship detected among Caucasian population (allele model,
OR = 1.08, 95% CI 0.92-1.26,
P = 0.352; dominant model,
OR = 1.05, 95% CI 0.87-1.26,
P = 0.639). The present meta-analysis had suggested
that Int7G24A polymorphism of gene TGFBR1 involved in the transforming growth factor
beta (TGF-β) signaling pathway had a significantly increased risk for
cancer development.
Collapse
|
6
|
Single Nucleotide Polymorphism (rs4932178) in the P1 Promoter of FURIN Is Not Prognostic to Colon Cancer. BIOMED RESEARCH INTERNATIONAL 2015; 2015:321276. [PMID: 26137475 PMCID: PMC4475562 DOI: 10.1155/2015/321276] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 01/18/2015] [Accepted: 01/26/2015] [Indexed: 01/05/2023]
Abstract
High expression of the proprotein processing enzyme FURIN has been associated with tumor progression and metastasis. A SNP (rs4932178) in the promoter of FURIN has been reported to affect expression in liver, with the T allele resulting in higher expression than the C allele. In this study we have investigated the association of this SNP with prognostic and biological subgroups of colorectal cancer (CRC). In a panel of 1382 patients with CRC, this SNP had no impact on overall survival or on postoperative risk of relapse. This SNP also could not be linked with FURIN expression levels in CRC samples from the patients. Furthermore, we demonstrate in luciferase reporter experiments in the colon cancer cell lines Caco-2 and SW480 and in the hepatocellular carcinoma cell line Huh 7 that expression is not affected by the SNP. Since, FURIN inhibition in human colon cancer cell lines has previously been shown to repress tumor metastases, association between FURIN gene expression levels and postoperative relapse-free survival was also investigated. However, no association could be found. Altogether, we could not confirm an effect of the SNP on FURIN expression in vitro and no correlations could be found in vivo with FURIN expression or outcome.
Collapse
|
7
|
Slattery ML, Lundgreen A. The influence of the CHIEF pathway on colorectal cancer-specific mortality. PLoS One 2014; 9:e116169. [PMID: 25541970 PMCID: PMC4277466 DOI: 10.1371/journal.pone.0116169] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 11/30/2014] [Indexed: 01/22/2023] Open
Abstract
Many components of the CHIEF (Convergence of Hormones, Inflammation, and Energy Related Factors) pathway could influence survival given their involvement in cell growth, apoptosis, angiogenesis, and tumor invasion stimulation. We used ARTP (Adaptive Rank Truncation Product) to test if genes in the pathway were associated with colorectal cancer-specific mortality. Colon cancer (n = 1555) and rectal cancer (n = 754) cases were followed over five years. Age, center, stage at diagnosis, and tumor molecular phenotype were considered when calculating ARTP p values. A polygenic risk score was used to summarize the magnitude of risk associated with this pathway. The JAK/STAT/SOC was significant for colon cancer survival (PARTP = 0.035). Fifteen genes (DUSP2, INFGR1, IL6, IRF2, JAK2, MAP3K10, MMP1, NFkB1A, NOS2A, PIK3CA, SEPX1, SMAD3, TLR2, TYK2, and VDR) were associated with colon cancer mortality (PARTP < 0.05); JAK2 (PARTP = 0.0086), PIK3CA (PARTP = 0.0098), and SMAD3 (PARTP = 0.0059) had the strongest associations. Over 40 SNPs were significantly associated with survival within the 15 significant genes (PARTP < 0.05). SMAD3 had the strongest association with survival (HRGG 2.46 95% CI 1.44,4.21 PTtrnd = 0.0002). Seven genes (IL2RA, IL8RA, IL8RB, IRF2, RAF1, RUNX3, and SEPX1) were significantly associated with rectal cancer (PARTP < 0.05). The HR for colorectal cancer-specific mortality among colon cancer cases in the upper at-risk alleles group was 11.81 (95% CI 7.07, 19. 74) and was 10.99 (95% CI 5.30, 22.78) for rectal cancer. These results suggest that several genes in the CHIEF pathway are important for colorectal cancer survival; the risk associated with the pathway merits validation in other studies.
Collapse
Affiliation(s)
- Martha L. Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, 383 Colorow Building, Salt Lake City, Utah, United States of America
- * E-mail:
| | - Abbie Lundgreen
- Department of Internal Medicine, University of Utah Health Sciences Center, 383 Colorow Building, Salt Lake City, Utah, United States of America
| |
Collapse
|
8
|
Ross JP, Lockett LJ, Tabor B, Saunders IW, Young GP, Macrae F, Blanco I, Capella G, Brown GS, Lockett TJ, Hannan GN. Little evidence for association between the TGFBR1*6A variant and colorectal cancer: a family-based association study on non-syndromic family members from Australia and Spain. BMC Cancer 2014; 14:475. [PMID: 24981199 PMCID: PMC4090415 DOI: 10.1186/1471-2407-14-475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 06/24/2014] [Indexed: 11/16/2022] Open
Abstract
Background Genome-wide linkage studies have identified the 9q22 chromosomal region as linked with colorectal cancer (CRC) predisposition. A candidate gene in this region is transforming growth factor β receptor 1 (TGFBR1). Investigation of TGFBR1 has focused on the common genetic variant rs11466445, a short exonic deletion of nine base pairs which results in truncation of a stretch of nine alanine residues to six alanine residues in the gene product. While the six alanine (*6A) allele has been reported to be associated with increased risk of CRC in some population based study groups this association remains the subject of robust debate. To date, reports have been limited to population-based case–control association studies, or case–control studies of CRC families selecting one affected individual per family. No study has yet taken advantage of all the genetic information provided by multiplex CRC families. Methods We have tested for an association between rs11466445 and risk of CRC using several family-based statistical tests in a new study group comprising members of non-syndromic high risk CRC families sourced from three familial cancer centres, two in Australia and one in Spain. Results We report a finding of a nominally significant result using the pedigree-based association test approach (PBAT; p = 0.028), while other family-based tests were non-significant, but with a p-value <; 0.10 in each instance. These other tests included the Generalised Disequilibrium Test (GDT; p = 0.085), parent of origin GDT Generalised Disequilibrium Test (GDT-PO; p = 0.081) and empirical Family-Based Association Test (FBAT; p = 0.096, additive model). Related-person case–control testing using the “More Powerful” Quasi-Likelihood Score Test did not provide any evidence for association (MQLS; p = 0.41). Conclusions After conservatively taking into account considerations for multiple hypothesis testing, we find little evidence for an association between the TGFBR1*6A allele and CRC risk in these families. The weak support for an increase in risk in CRC predisposed families is in agreement with recent meta-analyses of case–control studies, which estimate only a modest increase in sporadic CRC risk among 6*A allele carriers.
Collapse
Affiliation(s)
- Jason P Ross
- CSIRO Preventative Health Flagship, Sydney, NSW, Australia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Turpeinen H, Ortutay Z, Pesu M. Genetics of the first seven proprotein convertase enzymes in health and disease. Curr Genomics 2014; 14:453-67. [PMID: 24396277 PMCID: PMC3867721 DOI: 10.2174/1389202911314050010] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 09/13/2013] [Accepted: 09/14/2013] [Indexed: 12/16/2022] Open
Abstract
Members of the substilisin/kexin like proprotein convertase (PCSK) protease family cleave and convert immature pro-proteins into their biologically active forms. By cleaving for example prohormones, cytokines and cell membrane proteins, PCSKs participate in maintaining the homeostasis in a healthy human body. Conversely, erratic enzymatic function is thought to contribute to the pathogenesis of a wide variety of diseases, including obesity and hypercholestrolemia. The first characterized seven PCSK enzymes (PCSK1-2, FURIN, PCSK4-7) process their substrates at a motif made up of paired basic amino acid residues. This feature results in a variable degree of biochemical redundancy in vitro, and consequently, shared substrate molecules between the different PCSK enzymes. This redundancy has confounded our understanding of the specific biological functions of PCSKs. The physiological roles of these enzymes have been best illustrated by the phenotypes of genetically engineered mice and patients that carry mutations in the PCSK genes. Recent developments in genome-wide methodology have generated a large amount of novel information on the genetics of the first seven proprotein convertases. In this review we summarize the reported genetic alterations and their associated phenotypes.
Collapse
Affiliation(s)
- Hannu Turpeinen
- Immunoregulation, Institute of Biomedical Technology, University of Tampere, and BioMediTech, Tampere, Finland
| | - Zsuzsanna Ortutay
- Immunoregulation, Institute of Biomedical Technology, University of Tampere, and BioMediTech, Tampere, Finland
| | - Marko Pesu
- Immunoregulation, Institute of Biomedical Technology, University of Tampere, and BioMediTech, Tampere, Finland; ; Fimlab laboratories, Pirkanmaa Hospital District, Finland
| |
Collapse
|
10
|
Abstract
Neuromuscular diseases, which encompass disorders that affect muscle and its innervation, are highly heritable. Genetic diagnosis now frequently pinpoints the primary mutation responsible for a given neuromuscular disease. However, the results from genetic testing indicate that neuromuscular disease phenotypes may vary widely, even in individuals with the same primary disease-causing mutation. Clinical variability arises from both genetic and environmental factors. Genetic modifiers can now be identified using candidate gene as well as genomic approaches. The presence of genetic modifiers for neuromuscular disease helps define the clinical outcome and also highlights pathways of potential therapeutic utility. Herein, we will focus on single gene neuromuscular disorders, including muscular dystrophy, spinal muscular atrophy, and amyotrophic lateral sclerosis, and the methods that have been used to identify modifier genes. Animal models have been an invaluable resource for modifier gene discovery and subsequent mechanistic studies. Some modifiers, identified using animal models, have successfully translated to the human counterpart. Furthermore, in a few instances, modifier gene discovery has repetitively uncovered the same pathway, such as TGFβ signaling in muscular dystrophy, further emphasizing the relevance of that pathway. Knowledge of genetic factors that influence disease can have direct clinical applications for prognosis and predicted outcome.
Collapse
Affiliation(s)
- Kay-Marie Lamar
- Department of Human Genetics, Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL, USA
| | - Elizabeth M McNally
- Department of Human Genetics, Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
11
|
Ceco E, McNally EM. Modifying muscular dystrophy through transforming growth factor-β. FEBS J 2013; 280:4198-209. [PMID: 23551962 PMCID: PMC3731412 DOI: 10.1111/febs.12266] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Revised: 03/11/2013] [Accepted: 03/20/2013] [Indexed: 12/31/2022]
Abstract
Muscular dystrophy arises from ongoing muscle degeneration and insufficient regeneration. This imbalance leads to loss of muscle, with replacement by scar or fibrotic tissue, resulting in muscle weakness and, eventually, loss of muscle function. Human muscular dystrophy is characterized by a wide range of disease severity, even when the same genetic mutation is present. This variability implies that other factors, both genetic and environmental, modify the disease outcome. There has been an ongoing effort to define the genetic and molecular bases that influence muscular dystrophy onset and progression. Modifier genes for muscle disease have been identified through both candidate gene approaches and genome-wide surveys. Multiple lines of experimental evidence have now converged on the transforming growth factor-β (TGF-β) pathway as a modifier for muscular dystrophy. TGF-β signaling is upregulated in dystrophic muscle as a result of a destabilized plasma membrane and/or an altered extracellular matrix. Given the important biological role of the TGF-β pathway, and its role beyond muscle homeostasis, we review modifier genes that alter the TGF-β pathway and approaches to modulate TGF-β activity to ameliorate muscle disease.
Collapse
Affiliation(s)
- Ermelinda Ceco
- Committee on Cell Physiology, University of Chicago, IL 60637, USA
| | | |
Collapse
|
12
|
Flanigan KM, Ceco E, Lamar KM, Kaminoh Y, Dunn DM, Mendell JR, King WM, Pestronk A, Florence JM, Mathews KD, Finkel RS, Swoboda KJ, Gappmaier E, Howard MT, Day JW, McDonald C, McNally EM, Weiss RB. LTBP4 genotype predicts age of ambulatory loss in Duchenne muscular dystrophy. Ann Neurol 2013; 73:481-8. [PMID: 23440719 PMCID: PMC4106425 DOI: 10.1002/ana.23819] [Citation(s) in RCA: 188] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/02/2012] [Accepted: 11/17/2012] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) displays a clinical range that is not fully explained by the primary DMD mutations. Ltbp4, encoding latent transforming growth factor-β binding protein 4, was previously discovered in a genome-wide scan as a modifier of murine muscular dystrophy. We sought to determine whether LTBP4 genotype influenced DMD severity in a large patient cohort. METHODS We analyzed nonsynonymous single nucleotide polymorphisms (SNPs) from human LTBP4 in 254 nonambulatory subjects with known DMD mutations. These SNPs, V194I, T787A, T820A, and T1140M, form the VTTT and IAAM LTBP4 haplotypes. RESULTS Individuals homozygous for the IAAM LTBP4 haplotype remained ambulatory significantly longer than those heterozygous or homozygous for the VTTT haplotype. Glucocorticoid-treated patients who were IAAM homozygotes lost ambulation at 12.5 ± 3.3 years compared to 10.7 ± 2.1 years for treated VTTT heterozygotes or homozygotes. IAAM fibroblasts exposed to transforming growth factor (TGF) β displayed reduced phospho-SMAD signaling compared to VTTT fibroblasts, consistent with LTBP4' role as a regulator of TGFβ. INTERPRETATION LTBP4 haplotype influences age at loss of ambulation, and should be considered in the management of DMD patients.
Collapse
Affiliation(s)
- Kevin M. Flanigan
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
- Department of Neurology, The Ohio State University, Columbus, Ohio
| | - Ermelinda Ceco
- Department of Medicine, Department of Human Genetics, Committee on Cell Physiology, The University of Chicago
| | - Kay-Marie Lamar
- Department of Medicine, Department of Human Genetics, Committee on Cell Physiology, The University of Chicago
| | - Yuuki Kaminoh
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
| | - Diane M. Dunn
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, Utah
| | - Jerry R. Mendell
- The Center for Gene Therapy, Nationwide Children's Hospital, The Ohio State University, Columbus, Ohio
- Department of Pediatrics, The Ohio State University, Columbus, Ohio
- Department of Neurology, The Ohio State University, Columbus, Ohio
| | - Wendy M. King
- Department of Neurology, The Ohio State University, Columbus, Ohio
| | - Alan Pestronk
- Department of Neurology, Washington University at St. Louis, St. Louis, Missouri
| | - Julaine M. Florence
- Department of Neurology, Washington University at St. Louis, St. Louis, Missouri
| | - Katherine D. Mathews
- Department of Pediatrics and Neurology, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Richard S. Finkel
- Division of Neurology, The Children's Hospital of Philadelphia, and the Departments of Neurology and Pediatrics, Pearlman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Kathryn J. Swoboda
- Department of Neurology, University of Utah School of Medicine, University of Utah, Salt Lake City, Utah
| | - Eduard Gappmaier
- Department of Physical Therapy, University of Utah, Salt Lake City, Utah
| | - Michael T. Howard
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, Utah
| | - John W. Day
- Department of Neurology, University of Minnesota, Minneapolis, Minnesota
| | - Craig McDonald
- Department of Physical Medicine and Rehabilitation, University of California Davis, Sacramento, California
| | - Elizabeth M. McNally
- Department of Medicine, Department of Human Genetics, Committee on Cell Physiology, The University of Chicago
| | - Robert B. Weiss
- Department of Human Genetics, The University of Utah School of Medicine, Salt Lake City, Utah
| |
Collapse
|
13
|
Liu X, Shan Y, Xue B. Int7G24A polymorphism (rs334354) and cancer risk. Arch Med Sci 2013; 9:3-7. [PMID: 23515707 PMCID: PMC3598132 DOI: 10.5114/aoms.2013.33341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 03/19/2012] [Accepted: 07/09/2012] [Indexed: 02/05/2023] Open
Abstract
INTRODUCTION The transforming growth factor β (TGF-β) signaling system plays an important role in carcinogenesis. Alteration of TGF-β receptors is a potential mechanism in the development and progression of human cancers. Several studies have investigated the association between TGFBR1 gene Int7G24A and cancer risk, but the results are still inconclusive, so a meta-analysis is needed to verify the association. MATERIAL AND METHODS We carried out a literature search using the PubMed database (up to January 2012) to identify all papers that investigated the association between the Int7G24A polymorphism (rs334354) and cancer risk. The inclusion criteria were: (1) evaluation of the Int7G24A polymorphism and cancer risk, (2) case-control studies, (3) sufficient published data about genotype frequency. Also the strength of the association between Int7G24A polymorphism and cancer was measured by odds ratio (OR), which was calculated according to the method of Woolf. A χ(2)-based Q statistic test was performed to assess the between-study heterogeneity. RESULTS There are 10 studies including 2398 cases and 3465 controls in the research. Our results indicate that the TGFBR1 gene Int7G24A polymorphism is associated with cancer risk (A vs. G: OR = 1.35, 95% CI = 1.10-1.66, A/A+G/A vs. G/G: OR = 1.34, 95% CI = 1.05-1.72). CONCLUSIONS This meta-analysis suggests that the TGFBR1 gene Int7G24A polymorphism might be associated with an increased risk of cancer.
Collapse
Affiliation(s)
- Xiaolong Liu
- Urology Department, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | | | | |
Collapse
|
14
|
Association between TGFBR1 polymorphisms and cancer risk: a meta-analysis of 35 case-control studies. PLoS One 2012; 7:e42899. [PMID: 22905183 PMCID: PMC3414489 DOI: 10.1371/journal.pone.0042899] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2012] [Accepted: 07/12/2012] [Indexed: 12/15/2022] Open
Abstract
Background Numerous epidemiological studies have evaluated the association between TGFBR1 polymorphisms and the risk of cancer, however, the results remain inconclusive. To derive a more precise estimation of the relation, we conducted a comprehensive meta-analysis of all available case-control studies relating the TGFBR1*6A and IVS7+24G>A polymorphisms of the TGFBR1 gene to the risk of cancer. Methods Eligible studies were identified by search of electronic databases. Overall and subgroup analyses were performed. Odds ratio (OR) and 95% confidence interval (CI) were applied to assess the associations between TGFBR1*6A and IVS7+24G>A polymorphisms and cancer risk. Results A total of 35 studies were identified, 32 with 19,767 cases and 18,516 controls for TGFBR1*6A polymorphism and 12 with 4,195 cases and 4,383 controls for IVS7+24G>A polymorphism. For TGFBR1*6A, significantly elevated cancer risk was found in all genetic models (dominant OR = 1.11, 95% CI = 1.04∼1.18; recessive: OR = 1.36, 95% CI = 1.11∼1.66; additive: OR = 1.13, 95% CI = 1.05∼1.20). In subgroup analysis based on cancer type, increased cancer risk was found in ovarian and breast cancer. For IVS7+24G>A, significant correlation with overall cancer risk (dominant: OR = 1.39, 95% CI = 1.15∼1.67; recessive: OR = 2.23, 95% CI = 1.26∼3.92; additive: OR = 1.43, 95% CI = 1.14∼1.80) was found, especially in Asian population. In the subgroup analysis stratified by cancer type, significant association was found in breast and colorectal cancer. Conclusions Our investigations demonstrate that TGFBR1*6A and IVS7+24G>A polymorphisms of TGFBR1 are associated with the susceptibility of cancer, and further functional research should be performed to explain the inconsistent results in different ethnicities and cancer types.
Collapse
|
15
|
Liu Y, Zhou W, Zhong DW. Meta-analyses of the associations between four common TGF-β1 genetic polymorphisms and risk of colorectal tumor. Tumour Biol 2012; 33:1191-9. [PMID: 22396040 DOI: 10.1007/s13277-012-0364-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2012] [Accepted: 02/13/2012] [Indexed: 12/27/2022] Open
Abstract
The associations between four common genetic polymorphisms of transforming growth factor-β1 (TGF-β1 -509 C > T, +869 T > C, +915 G > C, and -800 G > A) and risk of colorectal tumor (including adenoma and cancer) have been widely studied. To date, no conclusions could be available because of controversial results reported. Thus, we conducted a meta-analysis to further assess the associations. We searched the databases of Medline, Embase, and Wangfang to identify eligible studies, and latest update was on January 1, 2012. Odds ratio (OR) and 95% confidence interval (95%CI) were calculated to present the associations. Our meta-analysis indicated that TGF-β1 -509 C > T, +869 T > C, +915 G > C, and -800 G > A were not associated with risk of colorectal adenoma (OR = 0.89 for C carriers vs. TT for -509 C > T, 1.03 for C carriers vs. TT for +869 T > C, 1.09 for C carriers vs. GG for +915 G > C, and 1.19 for A carriers vs. GG for 800 G > A). However, C allele of TGF-β1 -509 C > T and A allele of -800 G > A were associated with increased risk of colorectal cancer (CRC), and OR (95%CI) was 1.23 (0.99-1.52) for CC vs. TT for -509 C > T and 6.64 (3.46-12.72) for A carriers vs. GG. The positive association between -509 C allele and risk of CRC was more obvious when subgroup analyses were conducted for population-based and large sample-sized studies as well as Caucasians. In contrast, we did not observed any associations between TGF-β1 +869 T > C, +915 G > C, and risk of CRC. This study indicated that C allele of TGF-β1-509 C > T and A allele of -800 G > A might contribute to the increased risk of CRC, and could be used as two of genetic marks for screening individuals at high risk of CRC. Because of modest limitation, large sample-sized studies were required to confirm the findings.
Collapse
Affiliation(s)
- Yi Liu
- Department of Hepatobiliary Surgery,The Second Xiangya Hospital, Central South University, Renmin Road No.139, Changsha City, 410011 Hunan Province, China
| | | | | |
Collapse
|
16
|
Martinez-Canto A, Castillejo A, Mata-Balaguer T, Castillejo MI, Hernandez-Illan E, Irles E, Barbera VM, Egoavil C, Guarinos C, Alenda C, Ochoa E, Lazaro R, Fajardo S, Lacueva J, Calpena R, Soto JL. TGFBR1 intralocus epistatic interaction as a risk factor for colorectal cancer. PLoS One 2012; 7:e30812. [PMID: 22292045 PMCID: PMC3264637 DOI: 10.1371/journal.pone.0030812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2011] [Accepted: 12/21/2011] [Indexed: 11/24/2022] Open
Abstract
In colorectal cancer (CRC), an inherited susceptibility risk affects about 35% of patients, whereas high-penetrance germline mutations account for <6% of cases. A considerable proportion of sporadic tumors could be explained by the coinheritance of multiple low-penetrance variants, some of which are common. We assessed the susceptibility to CRC conferred by genetic variants at the TGFBR1 locus. We analyzed 14 polymorphisms and the allele-specific expression (ASE) of TGFBR1 in 1025 individuals from the Spanish population. A case-control study was undertaken with 504 controls and 521 patients with sporadic CRC. Fourteen polymorphisms located at the TGFBR1 locus were genotyped with the iPLEX Gold (MassARRAY-Sequenom) technology. Descriptive analyses of the polymorphisms and haplotypes and association studies were performed with the SNPator workpackage. No relevant associations were detected between individual polymorphisms or haplotypes and the risk of CRC. The TGFBR1*9A/6A polymorphism was used for the ASE analysis. Heterozygous individuals were analyzed for ASE by fragment analysis using cDNA from normal tissue. The relative level of allelic expression was extrapolated from a standard curve. The cutoff value was calculated with Youden's index. ASE was found in 25.4% of patients and 16.4% of controls. Considering both bimodal and continuous types of distribution, no significant differences between the ASE values of patients and controls were identified. Interestingly, a combined analysis of the polymorphisms and ASE for the association with CRC occurrence revealed that ASE-positive individuals carrying one of the most common haplotypes (H2: 20.7%) showed remarkable susceptibility to CRC (RR: 5.25; 95% CI: 2.547–5.250; p<0.001) with a synergy factor of 3.7. In our study, 54.1% of sporadic CRC cases were attributable to the coinheritance of the H2 haplotype and TGFBR1 ASE. These results support the hypothesis that the allelic architecture of cancer genes, rather than individual polymorphisms, more accurately defines the CRC risk.
Collapse
Affiliation(s)
| | - Adela Castillejo
- Molecular Genetics Laboratory, Elche University Hospital, Elche, Spain
| | | | | | | | - Esperanza Irles
- Molecular Genetics Laboratory, Elche University Hospital, Elche, Spain
| | | | - Cecilia Egoavil
- Molecular Genetics Laboratory, Elche University Hospital, Elche, Spain
| | - Carla Guarinos
- Molecular Genetics Laboratory, Elche University Hospital, Elche, Spain
| | - Cristina Alenda
- Department of Pathology, Alicante University Hospital, Alicante, Spain
| | - Enrique Ochoa
- Molecular Biopathology, Castellon Provincial Hospital, Castellon, Spain
| | - Rafael Lazaro
- Department of Pathology, La Plana Hospital, Villareal, Spain
| | - Silvia Fajardo
- Molecular Genetics Laboratory, Elche University Hospital, Elche, Spain
| | - Javier Lacueva
- Department of Surgery, Elche University Hospital, Elche, Spain
| | - Rafael Calpena
- Department of Surgery, Elche University Hospital, Elche, Spain
| | - Jose Luis Soto
- Molecular Genetics Laboratory, Elche University Hospital, Elche, Spain
- * E-mail:
| |
Collapse
|
17
|
Valle L. Debate about TGFBR1 and the susceptibility to colorectal cancer. World J Gastrointest Oncol 2012; 4:1-8. [PMID: 22347533 PMCID: PMC3277874 DOI: 10.4251/wjgo.v4.i1.1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2011] [Revised: 10/21/2011] [Accepted: 10/28/2011] [Indexed: 02/05/2023] Open
Abstract
Recent years have witnessed enormous progress in our understanding of the genetic predisposition to colorectal cancer (CRC). Estimates suggest that all or most genetic susceptibility mechanisms proposed so far, ranging from high-penetrance genes to low-risk alleles, account for about 60% of the population-attributable fraction of CRC predisposition. In this context, there is increasing interest in the gene encoding the transforming growth factor β receptor 1 (TGFBR1); first when over a decade ago a common polymorphism in exon 1 (rs11466445, TGFBR1*6A/9A) was suggested to be a risk allele for CRC, then when linkage studies identified the chromosomal region where the gene is located as susceptibility locus for familial CRC, and more recently when the allele-specific expression (ASE) of the gene was proposed as a risk factor for CRC. Published data on the association of TGFBR1 with CRC, regarding polymorphisms and ASE and including sporadic and familial forms of the disease, are often contradictory. This review gives a general overview of the most relevant studies in order to clarify the role of TGFBR1 in the field of CRC genetic susceptibility.
Collapse
Affiliation(s)
- Laura Valle
- Laura Valle, Hereditary Cancer Program, Catalan Institute of Oncology, IDIBELL, 08908 Hospitalet de Llobregat, Spain
| |
Collapse
|
18
|
Passarelli MN, Coghill AE, Hutter CM, Zheng Y, Makar KW, Potter JD, Newcomb PA. Common colorectal cancer risk variants in SMAD7 are associated with survival among prediagnostic nonsteroidal anti-inflammatory drug users: a population-based study of postmenopausal women. Genes Chromosomes Cancer 2011; 50:875-86. [PMID: 21910156 DOI: 10.1002/gcc.20913] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2011] [Accepted: 07/06/2011] [Indexed: 12/21/2022] Open
Abstract
Common single nucleotide polymorphisms (SNPs) in SMAD7 (18q21) have been linked to colorectal cancer (CRC) risk in genome-wide association studies, but little is known about their effects on survival. SMAD7 regulates gastrointestinal inflammation by inhibiting transforming growth factor-β (TGFB), which can act as both a tumor suppressor and a promoter of metastasis. Regular use of cyclooxygenase-2 (COX2) inhibitors, such as nonsteroidal anti-inflammatory drugs (NSAIDs), reduces the risk of developing CRC. Because COX2 overexpression reduces the growth suppressing effects of TGFB, we hypothesized that survival may depend on both SMAD7 genotype and prediagnostic NSAID use. Postmenopausal women, ages 50-74, diagnosed with incident invasive CRC from 1997 to 2002 were identified using the Seattle-Puget Sound Surveillance, Epidemiology, and End Results (SEER) cancer registry. Information on prediagnostic NSAID use and other risk factors was ascertained by interview, and women were followed-up for survival through December 31, 2009. Seven hundred and twenty-seven cases were genotyped for two GWAS hits in SMAD7 with minor allele frequency > 30%, one with minor allele associated with decreased risk (rs4939827) and one with minor allele associated with increased risk (rs4464148). Two hundred and forty-two deaths occurred, 160 attributable to CRC. Among those without distant disease at diagnosis, CRC-specific survival differed by genotype only for NSAID users: each minor allele of rs4939827 was associated with worse survival [hazard ratio (HR) = 2.67, 95% confidence interval (CI): 1.33-5.37] and each minor allele of rs4464148 was associated with better survival (HR = 0.41, CI 0.18-0.94). SMAD7 variants known to be important for CRC risk were associated with disease-specific survival among prediagnostic NSAID users.
Collapse
Affiliation(s)
- Michael N Passarelli
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | | | | | | | | | | |
Collapse
|
19
|
The association of polymorphisms on TGFBR1 and colorectal cancer risk: a meta-analysis. Mol Biol Rep 2011; 39:2567-74. [PMID: 21858550 DOI: 10.1007/s11033-011-1009-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2010] [Accepted: 06/02/2011] [Indexed: 01/06/2023]
Abstract
Epidemiological studies found inconsistent results on the association of two variants on TGFBR1 (TGFBR1*6A and Int7G24A) with colorectal cancer (CRC) risk. The present study was aimed to evaluate the association of these two variants with CRC susceptibility via the meta-analysis methods. For variant TGFBR1*6A, nine reports including 6,765 CRC patients and 8,496 unrelated controls were identified. The heterozygotes *6A/*9A showed a significant increased risk of CRC with the pooled OR was 1.12 (95% CI = 1.02-1.23), and the pooled OR for the homozygotes *6A/*6A was 1.13 (95% CI = 0.80-1.58) compared to the homozygotes *9A/*9A. However, under the dominant effect model, the TGFBR1*6A carriers showed a significantly increased CRC risk (pooled OR = 1.12, 95% CI = 1.03-1.23, *6A/*6A and *6A/*9A vs. *9A/*9A). For variant Int7G24A, three case-control studies with 1,074 cases and 1,945 controls were found. Although no significant association was found for heterozygosity Int7G24A carriers with CRC risk (pooled OR = 0.97, 95% CI = 0.67-1.42), the homozygosity A/A carriers showed a significant elevated risk of CRC (pooled OR = 1.68, 95% CI = 1.14-2.47) compared to G/G homozygotes. Under the recessive effect model, homozygotes A/A showed a 71% increase of CRC risk compared to the A/G and G/G genotype carriers (pooled OR = 1.71, 95% CI = 1.17-2.51). These data strongly suggested that the two polymorphisms of TGFBR1 may confer low-penetrance susceptibility of CRC risk.
Collapse
|
20
|
Slattery ML, Lundgreen A, Herrick JS, Wolff RK, Caan BJ. Genetic variation in the transforming growth factor-β signaling pathway and survival after diagnosis with colon and rectal cancer. Cancer 2011; 117:4175-83. [PMID: 21365634 DOI: 10.1002/cncr.26018] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2010] [Revised: 01/12/2011] [Accepted: 02/01/2011] [Indexed: 01/10/2023]
Abstract
BACKGROUND The transforming growth factor-β (TGF-β) signaling pathway is involved in many aspects of tumorigenesis, including angiogenesis and metastasis. The authors evaluated this pathway in association with survival after a diagnosis of colon or rectal cancer. METHODS The study included 1553 patients with colon cancer and 754 patients with rectal cancer who had incident first primary disease and were followed for a minimum of 7 years after diagnosis. Genetic variations were evaluated in the genes TGF-β1 (2 single nucleotide polymorphisms [SNPs]), TGF-β receptor 1 (TGF-βR1) (3 SNPs), smooth muscle actin/mothers against decapentaplegic homolog 1 (Smad1) (5 SNPs), Smad2 (4 SNPs), Smad3 (37 SNPs), Smad4 (2 SNPs), Smad7 (11 SNPs), bone morphogenetic protein 1 (BMP1) (11 SNPs), BMP2 (5 SNPs), BMP4 (3 SNPs), bone morphogenetic protein receptor 1A (BMPR1A) (9 SNPs), BMPR1B (21 SNPs), BMPR2 (11 SNPs), growth differentiation factor 10 (GDF10) (7 SNPs), Runt-related transcription factor 1 (RUNX1) (40 SNPs), RUNX2 (19 SNPs), RUNX3 (9 SNPs), eukaryotic translation initiation factor 4E (eiF4E) (3 SNPs), eukaryotic translation initiation factor 4E-binding protein 3 (eiF4EBP2) (2 SNPs), eiF4EBP3 (2 SNPs), and mitogen-activated protein kinase 1 (MAPK1) (6 SNPs). RESULTS After adjusting for American Joint Committee on Cancer stage and tumor molecular phenotype, 12 genes and 18 SNPs were associated with survival in patients with colon cancer, and 7 genes and 15 tagSNPs were associated with survival after a diagnosis of rectal cancer. A summary score based on "at-risk" genotypes revealed a hazard rate ratio of 5.10 (95% confidence interval, 2.56-10.15) for the group with the greatest number of "at-risk" genotypes; for rectal cancer, the hazard rate ratio was 6.03 (95% confidence interval, 2.83-12.75). CONCLUSIONS The current findings suggest that the presence of several higher risk alleles in the TGF-β signaling pathway increase the likelihood of dying after a diagnosis of colon or rectal cancer.
Collapse
Affiliation(s)
- Martha L Slattery
- Department of Internal Medicine, University of Utah Health Sciences Center, Salt Lake City, Utah 84108, USA.
| | | | | | | | | |
Collapse
|
21
|
Manne U, Shanmugam C, Katkoori VR, Bumpers HL, Grizzle WE. Development and progression of colorectal neoplasia. Cancer Biomark 2010; 9:235-65. [PMID: 22112479 PMCID: PMC3445039 DOI: 10.3233/cbm-2011-0160] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
A variety of genetic and molecular alterations underlie the development and progression of colorectal neoplasia (CRN). Most of these cancers arise sporadically due to multiple somatic mutations and genetic instability. Genetic instability includes chromosomal instability (CIN) and microsatellite instability (MSI), which is observed in most hereditary non-polyposis colon cancers (HNPCCs) and accounts for a small proportion of sporadic CRN. Although many biomarkers have been used in the diagnosis and prediction of the clinical outcomes of CRNs, no single marker has established value. New markers and genes associated with the development and progression of CRNs are being discovered at an accelerated rate. CRN is a heterogeneous disease, especially with respect to the anatomic location of the tumor, race/ethnicity differences, and genetic and dietary interactions that influence its development and progression and act as confounders. Hence, efforts related to biomarker discovery should focus on identification of individual differences based on tumor stage, tumor anatomic location, and race/ethnicity; on the discovery of molecules (genes, mRNA transcripts, and proteins) relevant to these differences; and on development of therapeutic approaches to target these molecules in developing personalized medicine. Such strategies have the potential of reducing the personal and socio-economic burden of CRNs. Here, we systematically review molecular and other pathologic features as they relate to the development, early detection, diagnosis, prognosis, progression, and prevention of CRNs, especially colorectal cancers (CRCs).
Collapse
Affiliation(s)
- Upender Manne
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, USA.
| | | | | | | | | |
Collapse
|