1
|
Qiao Q, Yuan SS, Shang J, Liu JX. Multi-View Enhanced Tensor Nuclear Norm and Local Constraint Model for Cancer Clustering and Feature Gene Selection. J Comput Biol 2023; 30:889-899. [PMID: 37471239 DOI: 10.1089/cmb.2023.0107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/22/2023] Open
Abstract
The analysis of cancer data from multi-omics can effectively promote cancer research. The main focus of this article is to cluster cancer samples and identify feature genes to reveal the correlation between cancers and genes, with the primary approach being the analysis of multi-view cancer omics data. Our proposed solution, the Multi-View Enhanced Tensor Nuclear Norm and Local Constraint (MVET-LC) model, aims to utilize the consistency and complementarity of omics data to support biological research. The model is designed to maximize the utilization of multi-view data and incorporates a nuclear norm and local constraint to achieve this goal. The first step involves introducing the concept of enhanced partial sum of tensor nuclear norm, which significantly enhances the flexibility of the tensor nuclear norm. After that, we incorporate total variation regularization into the MVET-LC model to further augment its performance. It enables MVET-LC to make use of the relationship between tensor data structures and sparse data while paying attention to the feature details of the tensor data. To tackle the iterative optimization problem of MVET-LC, the alternating direction method of multipliers is utilized. Through experimental validation, it is demonstrated that our proposed model outperforms other comparison models.
Collapse
Affiliation(s)
- Qian Qiao
- School of Computer Science, Qufu Normal University, Rizhao, China
| | - Sha-Sha Yuan
- School of Computer Science, Qufu Normal University, Rizhao, China
| | - Junliang Shang
- School of Computer Science, Qufu Normal University, Rizhao, China
| | - Jin-Xing Liu
- School of Computer Science, Qufu Normal University, Rizhao, China
| |
Collapse
|
2
|
Dang Y, Xu Q, Liu X, Wang L, Lin C. Clinical and functional characterisation of the SMAD4 germline variant c.1035C > A in a family with juvenile polyposis syndrome by whole-exome sequencing. Med Mol Morphol 2023; 56:78-83. [PMID: 35851413 DOI: 10.1007/s00795-022-00333-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/04/2022] [Indexed: 11/28/2022]
Abstract
Juvenile polyposis syndrome (JPS) is a rare autosomal dominant inherited disease characterised by multiple juvenile polyps. Genes with JPS-associated mutations and their correlation with the phenotype are currently unknown. Gastrointestinal endoscopy results of a 31-year-old female patient showed multiple polyps in the digestive tract, and the presence of juvenile polyps was confirmed by pathological examination. During follow-up, the patient underwent total gastrectomy and polypectomy several times. Five members of this family were diagnosed with JPS, of which two died and three survived. Full exon gene sequencing of eight members of this family revealed a SMAD4 (NM-005359.3) c.1035C > A (p.Cys345*) mutation. This mutation leads to premature codon termination, causing protein truncation. SMAD4 is a pathogenic gene associated with JPS. This is the first report of an association between the c.1035C > A mutation and JPS pathogenesis. Detection of JPS-related mutations in family members with a genetic predisposition for JPS is very important for genetic counselling, surgical intervention, long-term monitoring and follow-up, and drug treatment.
Collapse
Affiliation(s)
- Yuan Dang
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, China.,Innovation Center for Cancer Research, Fujian Medical University Cancer Hospital, Fujian Cancer Hospital, Fuzhou, Fujian, China.,Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fujian Cancer Hospital, Fuzhou, China
| | - Qianhui Xu
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, China.,Department of General Surgery, 900th hospital of Joint Logistics Support Force, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China.,Department of General Surgery, Dongfang Hospital of Xiamen university, School of Medicine, Xiamen University, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China
| | - Xiaofang Liu
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, China.,Department of General Surgery, 900th hospital of Joint Logistics Support Force, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China.,Department of General Surgery, Dongfang Hospital of Xiamen university, School of Medicine, Xiamen University, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China
| | - Lie Wang
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, China. .,Department of General Surgery, 900th hospital of Joint Logistics Support Force, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China. .,Department of General Surgery, Dongfang Hospital of Xiamen university, School of Medicine, Xiamen University, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China.
| | - Chen Lin
- Department of General Surgery, Fuzong Clinical Medical College, Fujian Medical University, Fuzhou, 350025, Fujian, China. .,Department of General Surgery, 900th hospital of Joint Logistics Support Force, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China. .,Department of General Surgery, Dongfang Hospital of Xiamen university, School of Medicine, Xiamen University, 156 Xierhuan Road, Fuzhou, 350025, Fujian, China.
| |
Collapse
|
3
|
Choi SH, Barker EC, Gerber KJ, Letterio JJ, Kim BG. Loss of p27Kip1 leads to expansion of CD4+ effector memory T cells and accelerates colitis-associated colon cancer in mice with a T cell lineage restricted deletion of Smad4. Oncoimmunology 2020; 9:1847832. [PMID: 33329939 PMCID: PMC7722707 DOI: 10.1080/2162402x.2020.1847832] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
The cyclin-dependent kinase inhibitor p27Kip1 is a tumor suppressor whose intrinsic activity in cancer cells correlates with tumor aggressiveness, invasiveness, and impaired tumor cell differentiation. Here we explore whether p27Kip1 indirectly influences tumor progression by restricting expansion and survival of effector memory T cell (TEM) populations in a preclinical model of spontaneous colitis-associated colorectal cancer (CAC). We show mRNA and protein expression of p27Kip1 to be significantly decreased in the colons of mice with a T cell-restricted deletion of the TGF-β intermediate, SMAD4 (Smad4TKO). Loss of p27Kip1 expression in T cells correlates with the onset of spontaneous CAC in Smad4TKO mice by 8 months of age. This phenotype is greatly accelerated by the introduction of a germline deletion of CDKN1b (the gene encoding p27Kip1) in Smad4TKO mice (Smad4TKO/p27Kip1-/-, DKO). DKO mice display colon carcinoma by 3 months of age and increased mortality compared to Smad4TKO. Importantly, the phenotype in DKO mice is associated with a significant increase in the frequency of effector CD4 T cells expressing abundant IFN-γ and with a concomitant decrease in Foxp3+ regulatory T cells, both in the intestinal mucosa and in the periphery. In addition, induction of inflammatory mediators (IFN-γ, TNF-γ, IL-6, IL-1β, iNOS) and activation of Stat1, Stat3, and IκB is also observed in the colon as early as 1–2 months of age. Our data suggest that genomic alterations known to influence p27Kip1 abundance in gastrointestinal cancers may indirectly promote epithelial malignancy by augmenting the production of inflammatory mediators from a spontaneously expanding pool of TEM cells.
Collapse
Affiliation(s)
- Sung Hee Choi
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - Emily C Barker
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA
| | - Kyle J Gerber
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio, USA
| | - John J Letterio
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA.,The Angie Fowler Adolescent and Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children's Hospital, Cleveland, Ohio, USA
| | - Byung-Gyu Kim
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH, USA.,Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| |
Collapse
|
4
|
MacFarland SP, Zelley K, Katona BW, Wilkins BJ, Brodeur GM, Mamula P. Gastrointestinal Polyposis in Pediatric Patients. J Pediatr Gastroenterol Nutr 2019; 69:273-280. [PMID: 31211762 PMCID: PMC7336255 DOI: 10.1097/mpg.0000000000002421] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gastrointestinal polyps are mucosal overgrowths that, if unchecked, can undergo malignant transformation. Although relatively uncommon in the pediatric age group, they can be the harbingers of multiorgan cancer risk and require close management and follow-up. Additionally, as many polyposis syndromes are inherited, appropriate genetic testing and management of relatives is vital for the health of the entire family. In this review, we discuss both common and uncommon childhood gastrointestinal polyposis syndromes in terms of clinical presentation, management, and surveillance. We also detail any additional malignancy risk and surveillance required in the pediatric age group (<21 years old). Through this review, we provide a framework for gastroenterologists to manage the multifaceted nature of pediatric polyposis syndromes.
Collapse
Affiliation(s)
- Suzanne P. MacFarland
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Kristin Zelley
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Bryson W. Katona
- Division of Gastroenterology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Benjamin J. Wilkins
- Department of Pathology and Laboratory Medicine, The Children’s Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Garrett M. Brodeur
- Division of Oncology, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| | - Petar Mamula
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA
| |
Collapse
|
5
|
Karlsson T, Cherif H. Mutations in the ENG, ACVRL1, and SMAD4 genes and clinical manifestations of hereditary haemorrhagic telangiectasia: experience from the Center for Osler's Disease, Uppsala University Hospital. Ups J Med Sci 2018; 123:153-157. [PMID: 30251589 PMCID: PMC6198721 DOI: 10.1080/03009734.2018.1483452] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
AIM The aim of this retrospective single-centre study was to evaluate whether mutations in the ENG, ACVRL1, and SMAD4 genes were associated with different phenotypes in hereditary haemorrhagic telangiectasia (HHT). METHODS The case records of 21 HHT patients with verified mutations in ENG, ACVRL1, or SMAD4 genes were reviewed. The numbers of HHT diagnostic criteria fulfilled for the three genotypes were compared, as was the prevalence of complications such as iron deficiency anaemia, gastrointestinal haemorrhage, stroke, and cerebral abscess. RESULTS Our results indicate that mutations in the ENG (HHT1), ACVRL1 (HHT2), and SMAD4 genes result in different HHT phenotypes. Epistaxis debuts earlier and may be more severe in HHT1 than in HHT2. The prevalence of pulmonary arteriovenous malformations (AVM) is higher in HHT type 1, whereas hepatic AVMs are more common in HHT2. One patient with mutations in both ENG and ACVRL1 genes was identified, as were two SMAD4-mutated patients suffering from the overlapping juvenile polyposis-HHT syndrome. Nearly one in five patients in our HHT population has been diagnosed with stroke or cerebral abscess, indicating a high prevalence of cerebral complications. CONCLUSION Our results showing that ENG and ACVRL1 gene mutations result in different HHT phenotypes confirm the results from other HHT centres worldwide. Cerebral complications of HHT are common, underscoring the importance of regular screening for pulmonary AVMs and early intervention against such AVMs. We have identified an HHT patient with simultaneous mutations in the ENG and ACVRL1 genes. Surprisingly, this patient has had a mild course of the disease.
Collapse
Affiliation(s)
- Torbjörn Karlsson
- Department of Haematology, Uppsala University Hospital, Uppsala, Sweden
- Center for Osler’s Disease, Uppsala University Hospital, Uppsala, Sweden
- Department of Medicine, Västmanlands Hospital, Västerås, Sweden
- CONTACT Torbjörn Karlsson Department of Haematology, Uppsala University Hospital, 751 85Uppsala, Sweden
| | - Honar Cherif
- Department of Haematology, Uppsala University Hospital, Uppsala, Sweden
- Center for Osler’s Disease, Uppsala University Hospital, Uppsala, Sweden
| |
Collapse
|
6
|
Barker EC, Kim BG, Yoon JH, Tochtrop GP, Letterio JJ, Choi SH. Potent suppression of both spontaneous and carcinogen-induced colitis-associated colorectal cancer in mice by dietary celastrol supplementation. Carcinogenesis 2018; 39:36-46. [PMID: 29069290 PMCID: PMC5862246 DOI: 10.1093/carcin/bgx115] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 09/22/2017] [Accepted: 10/20/2017] [Indexed: 12/16/2022] Open
Abstract
Celastrol is an anti-inflammatory natural triterpenoid, isolated from the herb Tripterygium wilfordii or thunder god vine. Here, we define mechanisms mediating anti-inflammatory activity of celastrol and demonstrate efficacy of a dietary celastrol supplement for chemoprevention of inflammation-driven carcinogenesis in mice. Dietary celastrol (31.25 ppm in rodent diet from 8 weeks to 25 weeks of age) is well tolerated and protects against LPS-induced acute inflammation in C57BL/6 mice, potently suppressing LPS-induction of inducible nitric oxide synthase (iNOS), cyclooxygenase (COX)-2, Interleukin (IL)-6 and IL-1β. To test whether dietary celastrol suppresses inflammation-driven colorectal cancer (CRC), we employed a unique model of spontaneous, inflammation-driven CRC in mice harboring a germ line deletion of the p27Kip1 gene and a T cell-specific deletion of Smad4 gene (Smad4co/co;Lck-crep27Kip1-/-or DKO), which develop severe intestinal inflammation and carcinogenesis as early as 3 months of age. Exposure of DKO mice to daily dietary celastrol (12.5 ppm in diet) from 6 weeks of age significantly suppressed development of colitis-associated CRC (CAC). Celastrol chemoprevention of CAC in this new model of intestinal neoplasia was associated with significant suppression of iNOS at 4 months of age, and iNOS, COX-2 and NFκB at 6 months of age, with significant reduction in inflammatory cytokines, IL-6 and IL-1β. Chemoprevetion of CAC by dietary celastrol was further confirmed in the model of azoxymethane (AOM) plus dextran sodium sulfate (DSS)-induced carcinogenesis in C57BL/6 mice. These data suggest the potential for celastrol as a safe and effective dietary supplement in the chemoprevention of CAC in humans.
Collapse
Affiliation(s)
- Emily C Barker
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio
| | - Byung-Gyu Kim
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| | - Ji Hee Yoon
- University of Notre Dame, Notre Dame, Indiana
| | - Gregory P Tochtrop
- Department of Chemistry, Case Western Reserve University, Cleveland, Ohio
- The Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
| | - John J Letterio
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
- The Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio
- The Angie Fowler Adolescent & Young Adult Cancer Institute, University Hospitals Rainbow Babies & Children’s Hospital, Cleveland, Ohio
| | - Sung Hee Choi
- Department of Pediatrics, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
7
|
Burmester JK, Bell LN, Cross D, Meyer P, Yale SH. A SMAD4 mutation indicative of juvenile polyposis syndrome in a family previously diagnosed with Menetrier's disease. Dig Liver Dis 2016; 48:1255-9. [PMID: 27375208 DOI: 10.1016/j.dld.2016.06.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 05/31/2016] [Accepted: 06/09/2016] [Indexed: 12/11/2022]
Abstract
BACKGROUND Menetrier's disease (MD) is a rare disease with unknown aetiology, characterized by hypertrophic folds within the fundus and body of the stomach. AIMS We investigated mutations of the candidate genes SMAD4, BMPR1A, TGF-α, and PDX1 within a family with MD. METHODS A large 4-generation family with MD was identified. This family had 5 cases of MD, 1 case of MD and juvenile polyposis syndrome (JPS) and 3 cases of JPS. Participants provided saliva for DNA extraction and completed a health questionnaire designed to assess conditions that may be found in patients with MD. Following pedigree analysis, we sequenced the coding regions of the SMAD4 and BMPR1A genes and the regulatory regions of the TGF-α and PDX1 genes in affected and non-affected family members. RESULTS No mutations were identified in the sequenced regions of BMPR1A, TGF-α, or PDX1. A dominant 1244_1247delACAG mutation of SMAD4 was identified in each of the subjects with JPS as well as in each of the subjects with MD. Although this mutation segregated with disease, there were also unaffected/undiagnosed carriers. CONCLUSION The 1244_1247delACAG mutation of SMAD4 is the cause of JPS and the likely cause of MD in a large family initially diagnosed with MD.
Collapse
Affiliation(s)
- James K Burmester
- Department of Research, Gundersen Medical Foundation, LaCrosse, WI, United States
| | - Lauren N Bell
- Department of Internal Medicine and Graduate Medical Education, North Florida Regional Medical Center, Gainesville, FL, United States
| | - Deanna Cross
- Office of Research Support, Christus Health, Irving, TX, United States
| | - Patrick Meyer
- University of Wisconsin Hospital and Clinics, Madison, WI, United States
| | - Steven H Yale
- Department of Internal Medicine and Graduate Medical Education, North Florida Regional Medical Center, Gainesville, FL, United States.
| |
Collapse
|
8
|
A Novel SMAD4 Mutation Causing Severe Juvenile Polyposis Syndrome with Protein Losing Enteropathy, Immunodeficiency, and Hereditary Haemorrhagic Telangiectasia. Case Rep Gastrointest Med 2015; 2015:140616. [PMID: 25705527 PMCID: PMC4331480 DOI: 10.1155/2015/140616] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 01/16/2015] [Accepted: 01/20/2015] [Indexed: 12/17/2022] Open
Abstract
Juvenile polyposis syndrome (JPS) is a rare genetic disorder characterized by juvenile polyps of the gastrointestinal tract. We present a new pathogenic mutation of the SMAD4 gene and illustrate the need for a multidisciplinary health care approach to facilitate the correct diagnosis. The patient, a 47-year-old Caucasian woman, was diagnosed with anaemia at the age of 12. During the following 30 years, she developed numerous gastrointestinal polyps. The patient underwent several operations, and suffered chronic abdominal pain, malnutrition, and multiple infections. Screening of the SMAD4 gene revealed a novel, disease-causing mutation. In 2012, the patient suffered hypoalbuminemia and a large polyp in the small bowel was found. Gamma globulin was given but the patient responded with fever and influenza-like symptoms and refused more treatment. The patient underwent surgery in 2014 and made an uneventful recovery. At follow-up two months later albumin was 38 g/L and IgG was 6.9 g/L. Accurate diagnosis is essential for medical care. For patients with complex symptomatology, often with rare diseases, this is best provided by multidisciplinary teams including representatives from clinical genetics. Patients with a SMAD4 mutation should be followed up both for JPS and haemorrhagic hereditary telangiectasia and may develop protein loosing enteropathy and immunodeficiency.
Collapse
|
9
|
Cichy W, Klincewicz B, Plawski A. Juvenile polyposis syndrome. Arch Med Sci 2014; 10:570-7. [PMID: 25097590 PMCID: PMC4107262 DOI: 10.5114/aoms.2014.43750] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 08/01/2011] [Accepted: 10/21/2011] [Indexed: 12/13/2022] Open
Abstract
Juvenile polyposis syndrome (JPS) is an autosomal dominant predisposition to the occurrence of hamartomatous polyps in the gastrointestinal tract. Diagnosis of JPS is based on the occurrence of numerous colon and rectum polyps or any number of polyps with family history and, in the case of juvenile polyps, their occurrence also outside the large intestine. The JPS is caused by mutations in SMAD4 and BMPR1A. Products of the SMAD4 gene are involved in signal transduction in the transforming growth factor β pathway and BMPR1A protein is a receptor belonging to the family of transmembrane serine/threonine kinases. Both proteins are responsible for processes determining appropriate development of colonic mucosa. The JPS belongs to the group of hamartomatous polyposes. The hamartomatous polyposis syndromes constitute a group of diseases in which manifestations differ slightly and only molecular diagnostics gives the possibility of verifying the clinical diagnosis.
Collapse
Affiliation(s)
- Wojciech Cichy
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Beata Klincewicz
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Andrzej Plawski
- Institute of Human Genetics, Polish Academy of Sciences, Poznan, Poland
| |
Collapse
|
10
|
Wain KE, Ellingson MS, McDonald J, Gammon A, Roberts M, Pichurin P, Winship I, Riegert-Johnson DL, Weitzel JN, Lindor NM. Appreciating the broad clinical features of SMAD4 mutation carriers: a multicenter chart review. Genet Med 2014; 16:588-93. [PMID: 24525918 DOI: 10.1038/gim.2014.5] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Accepted: 01/06/2014] [Indexed: 12/18/2022] Open
Abstract
Heterozygous loss-of-function SMAD4 mutations are associated with juvenile polyposis syndrome and hereditary hemorrhagic telangiectasia. Some carriers exhibit symptoms of both conditions, leading to juvenile polyposis-hereditary hemorrhagic telangiectasia syndrome. Three families have been reported with connective tissue abnormalities. To better understand the spectrum and extent of clinical findings in SMAD4 carriers, medical records of 34 patients (20 families) from five clinical practices were reviewed. Twenty-one percent of the patients (7/34) had features suggesting a connective tissue defect: enlarged aortic root (n = 3), aortic and mitral insufficiency (n = 2), aortic dissection (n = 1), retinal detachment (n = 1), brain aneurysms (n = 1), and lax skin and joints (n = 1). Juvenile polyposis-specific findings were almost uniformly present but variable. Ninety-seven percent of the patients had colon polyps that were generally pan-colonic and of variable histology and number. Forty-eight percent of the patients (15/31) had extensive gastric polyposis. Hereditary hemorrhagic telangiectasia features, including epistaxis (19/31, 61%), mucocutaneous telangiectases (15/31, 48%), liver arteriovenous malformation (6/16, 38%), brain arteriovenous malformation (1/26, 4%), pulmonary arteriovenous malformation (9/17, 53%), and intrapulmonary shunting (14/23, 61%), were documented in 76% of the patients. SMAD4 carriers should be managed for juvenile polyposis and hereditary hemorrhagic telangiectasia because symptoms of both conditions are likely yet unpredictable. Connective tissue abnormalities are an emerging component of juvenile polyposis-hereditary hemorrhagic telangiectasia syndrome, and larger studies are needed to understand these manifestations.
Collapse
Affiliation(s)
- Karen E Wain
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Jamie McDonald
- Department of Pathology, University of Utah, Salt Lake City, Utah, USA
| | - Amanda Gammon
- High Risk Cancer Clinics, Huntsman Cancer Institute, Salt Lake City, Utah, USA
| | | | - Pavel Pichurin
- Medical Genetics, Mayo Clinic, Rochester, Minnesota, USA
| | - Ingrid Winship
- 1] Department of Medicine, University of Melbourne, Melbourne, Australia [2] Genetic Medicine, Royal Melbourne Hospital, Melbourne, Australia
| | | | | | | |
Collapse
|
11
|
Jee MJ, Yoon SM, Kim EJ, Choi HJ, Kim JW, Sung RH, Han JH, Chae HB, Park SM, Youn SJ. A novel germline mutation in exon 10 of the SMAD4 gene in a familial juvenile polyposis. Gut Liver 2013; 7:747-51. [PMID: 24312718 PMCID: PMC3848546 DOI: 10.5009/gnl.2013.7.6.747] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Revised: 05/28/2013] [Accepted: 05/28/2013] [Indexed: 12/21/2022] Open
Abstract
Familial juvenile polyposis (FJP) is a rare autosomal dominant hereditary disorder that is characterized by the development of multiple distinct juvenile polyps in the gastrointestinal tract and an increased risk of cancer. Recently, germline mutations, including mutations in the SMAD4, BMPR1A, PTEN and, possibly, ENG genes, have been found in patients with juvenile polyps. We herein report a family with juvenile polyposis syndrome (JPS) with a novel germline mutation in the SMAD4 gene. A 21-year-old man presented with rectal bleeding and was found to have multiple polyps in his stomach, small bowel, and colon. His mother had a history of gastrectomy for multiple gastric polyps with anemia and a history of colectomy for colon cancer. A review of the histology of the polyps revealed juvenile polyps in both patients. Subsequently, mutation screening in DNA samples from the patients revealed a germline mutation in the SMAD4 gene. The pair had a novel mutation in exon 10 (stop codon at tyrosine 413). To our knowledge, this mutation has not been previously described. Careful family history collection and genetic screening in JPS patients are needed to identify FJP, and regular surveillance is recommended.
Collapse
Affiliation(s)
- Myung Jin Jee
- Department of Internal Medicine, Chungbuk National University Hospital, Chungbuk National University College of Medicine, Cheongju, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Piepoli A, Mazzoccoli G, Panza A, Tirino V, Biscaglia G, Gentile A, Valvano MR, Clemente C, Desiderio V, Papaccio G, Bisceglia M, Andriulli A. A unifying working hypothesis for juvenile polyposis syndrome and Ménétrier's disease: specific localization or concomitant occurrence of a separate entity? Dig Liver Dis 2012; 44:952-6. [PMID: 22748914 DOI: 10.1016/j.dld.2012.05.019] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Revised: 05/14/2012] [Accepted: 05/24/2012] [Indexed: 12/11/2022]
Abstract
BACKGROUND Juvenile polyposis syndrome with gastric involvement may mimic Ménétrier's disease, which is correlated to transforming growth factor (TGF)α overproduction and PDX1 upregulation in the gastric fundus. AIM We report a family with juvenile polyposis syndrome where one member showed typical features of Ménétrier's disease and concomitant Helicobacter pylori infection. METHODS We studied a 31-year-old woman belonging to a family with juvenile polyposis syndrome, who exhibited a particular form of hyperplastic gastropathy diagnosed as Ménétrier's disease with Helicobacter pylori infection. RESULTS TGFα overexpression and undetectable PDX1 expression were demonstrated in the fundic gastric biopsy specimens. In all affected members of the family we identified a 4-bp deletion in exon 9 of SMAD4 gene, a mutation usually associated with a more virulent form of juvenile polyposis syndrome with a higher incidence of gastric and colonic polyposis. CONCLUSION To explain the association of juvenile polyposis syndrome with Ménétrier's disease we hypothesized a new mechanism that involves TGFβ-SMAD4 pathway inactivation and TGFα overexpression related to Helicobacter pylori infection.
Collapse
Affiliation(s)
- Ada Piepoli
- Laboratory of Gastroenterology, Scientific Institute and Regional General Hospital Casa Sollievo della Sofferenza, S. Giovanni Rotondo, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Chapnick DA, Warner L, Bernet J, Rao T, Liu X. Partners in crime: the TGFβ and MAPK pathways in cancer progression. Cell Biosci 2011; 1:42. [PMID: 22204556 PMCID: PMC3275500 DOI: 10.1186/2045-3701-1-42] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2011] [Accepted: 12/28/2011] [Indexed: 12/27/2022] Open
Abstract
The TGFβ and Ras-MAPK pathways play critical roles in cell development and cell cycle regulation, as well as in tumor formation and metastasis. In the absence of cellular transformation, these pathways operate in opposition to one another, where TGFβ maintains an undifferentiated cell state and suppresses proliferation, while Ras-MAPK pathways promote proliferation, survival and differentiation. However, in colorectal and pancreatic cancers, the opposing pathways' mechanisms are simultaneously activated in order to promote cancer progression and metastasis. Here, we highlight the roles of the TGFβ and Ras-MAPK pathways in normal and malignant states, and provide an explanation for how the concomitant activation of these pathways drives tumor biology. Finally, we survey potential therapeutic targets in these pathways.
Collapse
Affiliation(s)
- Douglas A Chapnick
- Department of Chemistry and Biochemistry and Molecular, Cellular and Developmental Biology
| | - Lisa Warner
- Department of Chemistry and Biochemistry and Molecular, Cellular and Developmental Biology
| | | | - Timsi Rao
- Department of Chemistry and Biochemistry and Molecular, Cellular and Developmental Biology
| | - Xuedong Liu
- Department of Chemistry and Biochemistry and Molecular, Cellular and Developmental Biology
| |
Collapse
|
14
|
Du LY, Chen MS, Liu QF, Cheng XL, Li XM. Treatment with Compound Indigo Granules down-regulates NF-κB P65 and TNF-α expression in ulcerative colitis in rats. Shijie Huaren Xiaohua Zazhi 2011; 19:1290-1294. [DOI: 10.11569/wcjd.v19.i12.1290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To determine the mechanism underlying the therapeutic effects of Compound Indigo Granules (CIG) against ulcerative colitis by investigating the effect of treatment with CIG on the expression of nuclear factor-κB P65 (NF-κB P65) and tumor necrosis factor (TNF)-α in the colon of rats with experimental ulcerative colitis.
METHODS: Ulcerative colitis was induced in rats with trinitrobenzenesulfonic acid (TNBS). Rats were divided into control group, model group, salazosulfapyridine (SASP) group, low-, medium-, and high-dose CIG groups. Except the control group, the other groups were intragastrically given normal saline, SASP, and different doses of CIG from day 3 after model induction for 10 days, respectively. On day 14, all rats were killed to take colon and serum samples for measuring colonic NF-κB P65 expression by immunohistochemisty and serum TNF-α levels by enzyme-linked immunosorbent assay (ELISA).
RESULTS: The levels of NF-κB P65 protein expression in the colon and serum TNF-α were significantly higher in the model group than in the control group (0.138 ± 0.003 vs 0.276 ± 0.0081; 18.990 ± 3.964 vs 67.657 ± 3.580, both P < 0.05) but were significantly lower in the high-dose CIG group than in the model group (0.217 ± 0.007 vs 0.276 ± 0.008; 27.783 ± 2.867 vs 67.657 ± 3.580, both P < 0.05).
CONCLUSION: Treatment with CIG significantly decreased the levels of NF-κB P65 protein expression in the colon and serum TNF-α in rats with ulcerative colitis. CIG exerts therapeutic effects against ulcerative colitis possibly via mechanisms associated with the NF-κB signaling pathway and down-regulation of TNF-α.
Collapse
|
15
|
Gallione C, Aylsworth AS, Beis J, Berk T, Bernhardt B, Clark RD, Clericuzio C, Danesino C, Drautz J, Fahl J, Fan Z, Faughnan ME, Ganguly A, Garvie J, Henderson K, Kini U, Leedom T, Ludman M, Lux A, Maisenbacher M, Mazzucco S, Olivieri C, Ploos van Amstel JK, Prigoda-Lee N, Pyeritz RE, Reardon W, Vandezande K, Waldman JD, White RI, Williams CA, Marchuk DA. Overlapping spectra of SMAD4 mutations in juvenile polyposis (JP) and JP-HHT syndrome. Am J Med Genet A 2010; 152A:333-9. [PMID: 20101697 DOI: 10.1002/ajmg.a.33206] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Juvenile polyposis (JP) and hereditary hemorrhagic telangiectasia (HHT) are clinically distinct diseases caused by mutations in SMAD4 and BMPR1A (for JP) and endoglin and ALK1 (for HHT). Recently, a combined syndrome of JP-HHT was described that is also caused by mutations in SMAD4. Although both JP and JP-HHT are caused by SMAD4 mutations, a possible genotype:phenotype correlation was noted as all of the SMAD4 mutations in the JP-HHT patients were clustered in the COOH-terminal MH2 domain of the protein. If valid, this correlation would provide a molecular explanation for the phenotypic differences, as well as a pre-symptomatic diagnostic test to distinguish patients at risk for the overlapping but different clinical features of the disorders. In this study, we collected 19 new JP-HHT patients from which we identified 15 additional SMAD4 mutations. We also reviewed the literature for other reports of JP patients with HHT symptoms with confirmed SMAD4 mutations. Our combined results show that although the SMAD4 mutations in JP-HHT patients do show a tendency to cluster in the MH2 domain, mutations in other parts of the gene also cause the combined syndrome. Thus, any mutation in SMAD4 can cause JP-HHT. Any JP patient with a SMAD4 mutation is, therefore, at risk for the visceral manifestations of HHT and any HHT patient with SMAD4 mutation is at risk for early onset gastrointestinal cancer. In conclusion, a patient who tests positive for any SMAD4 mutation must be considered at risk for the combined syndrome of JP-HHT and monitored accordingly.
Collapse
Affiliation(s)
- Carol Gallione
- Duke University Medical Center, Durham, North Carolina 27710, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Yang G, Yang X. Smad4-mediated TGF-beta signaling in tumorigenesis. Int J Biol Sci 2010; 6:1-8. [PMID: 20087440 PMCID: PMC2808050 DOI: 10.7150/ijbs.6.1] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2009] [Accepted: 12/23/2009] [Indexed: 12/12/2022] Open
Abstract
Transforming growth factor-β (TGF-β) family members exert their function via specific type I and type II serine/threonine kinase receptors and intracellular Smad transcription factors, including the common mediator Smad4. The dual effects of TGF-β signaling on tumor initiation and progression are cell-specific and yet to be determined under distinct contexts. A number of genetically manipulated mouse models with alterations in the TGF-β pathway genes, particularly the pivotal Smad4, revealed that these genes play crucial functions in maintaining tissue homeostasis and suppressing tumorigenesis. Loss of Smad4 plays a causal role in initiating squamous cell carcinomas of skin and upper digestive tract as well as adenocarcinomas of gastrointestinal tract. However, for some cancers like pancreatic and cholangiocellular carcinomas, Smad4 deficiency does not initiate the tumorigenesis but acts as a promoter to accelerate or synergize the development and progression of cancers that are started by other oncogenic pathways. Intriguingly, emerging evidences from mouse models have highlighted the important roles of non-cell autonomous effects of Smad4-mediated TGF-β signaling in the inhibition of oncogenesis. All these data have greatly deepened our understanding of molecular mechanisms of cell-autonomous and non-cell autonomous effect of Smad4-mediated TGF-β signaling in suppressing carcinogenesis, which may facilitate the development of successful therapies targeting TGF-β signaling for the treatment of human cancers.
Collapse
Affiliation(s)
- Guan Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, AMMS, Beijing, P.R. China
| | | |
Collapse
|
17
|
Abstract
Transforming growth factor-beta (TGF-beta) family members exert their function via specific type I and type II serine/threonine kinase receptors and intracellular Smad transcription factors, including the common mediator Smad4. The dual effects of TGF-beta signaling on tumor initiation and progression are cell-specific and yet to be determined under distinct contexts. A number of genetically manipulated mouse models with alterations in the TGF-beta pathway genes, particularly the pivotal Smad4, revealed that these genes play crucial functions in maintaining tissue homeostasis and suppressing tumorigenesis. Loss of Smad4 plays a causal role in initiating squamous cell carcinomas of skin and upper digestive tract as well as adenocarcinomas of gastrointestinal tract. However, for some cancers like pancreatic and cholangiocellular carcinomas, Smad4 deficiency does not initiate the tumorigenesis but acts as a promoter to accelerate or synergize the development and progression of cancers that are started by other oncogenic pathways. Intriguingly, emerging evidences from mouse models have highlighted the important roles of non-cell autonomous effects of Smad4-mediated TGF-beta signaling in the inhibition of oncogenesis. All these data have greatly deepened our understanding of molecular mechanisms of cell-autonomous and non-cell autonomous effect of Smad4-mediated TGF-beta signaling in suppressing carcinogenesis, which may facilitate the development of successful therapies targeting TGF-beta signaling for the treatment of human cancers.
Collapse
Affiliation(s)
- Guan Yang
- State Key Laboratory of Proteomics, Genetic Laboratory of Development and Diseases, Institute of Biotechnology, AMMS, Beijing, P.R. China
| | | |
Collapse
|
18
|
Calva-Cerqueira D, Chinnathambi S, Pechman B, Bair J, Larsen-Haidle J, Howe JR. The rate of germline mutations and large deletions of SMAD4 and BMPR1A in juvenile polyposis. Clin Genet 2008; 75:79-85. [PMID: 18823382 DOI: 10.1111/j.1399-0004.2008.01091.x] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Juvenile polyposis (JPS) is an autosomal dominant syndrome that predisposes individuals to develop gastrointestinal polyps and cancer. Germline point mutations in SMAD4 and BMPR1A have been identified as causing JPS in approximately 40-60% of patients, but few studies have looked at the rate of large deletions. In this study, we determined the overall prevalence of genetic changes of SMAD4 and BMPR1A by sequencing and by screening for larger deletions. DNA was extracted from 102 JPS probands, and each exon and intron-exon boundary of SMAD4 and BMPR1A were sequenced. Coding and non-coding exons of SMAD4 and BMPR1A were screened for deletions with multiplex ligation-dependent probe amplification (MLPA). By sequencing, 20 probands had point mutations of SMAD4 and 22 of BMPR1A. By MLPA, one proband had deletion of most of SMAD4, one of both BMPR1A and PTEN, one of the 5' end of BMPR1A, and another of the 5' end of SMAD4. The overall prevalence of SMAD4 and BMPR1A point mutations and deletions in JPS was 45% in the largest series of patients to date. Large deletions are less frequent in JPS patients, but represent other heritable causes of JPS, which should be screened for in pre-symptomatic genetic testing.
Collapse
Affiliation(s)
- D Calva-Cerqueira
- Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA
| | | | | | | | | | | |
Collapse
|
19
|
Pintiliciuc O, Heresbach D, de-Lajarte-Thirouard AS, Dugast C, Reignier A, Cottereau J, Bretagne JF. Gastric involvement in juvenile polyposis associated with germline SMAD4 mutations: An entity characterized by a mixed hypertrophic and polypoid gastropathy. ACTA ACUST UNITED AC 2008; 32:445-50. [DOI: 10.1016/j.gcb.2007.11.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2007] [Accepted: 11/11/2007] [Indexed: 11/30/2022]
|
20
|
|
21
|
Ball EV, Stenson PD, Abeysinghe SS, Krawczak M, Cooper DN, Chuzhanova NA. Microdeletions and microinsertions causing human genetic disease: common mechanisms of mutagenesis and the role of local DNA sequence complexity. Hum Mutat 2006; 26:205-13. [PMID: 16086312 DOI: 10.1002/humu.20212] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
In the Human Gene Mutation Database (www.hgmd.org), microdeletions and microinsertions causing inherited disease (both defined as involving < or = 20 bp of DNA) account for 8,399 (17%) and 3,345 (7%) logged mutations, in 940 and 668 genes, respectively. A positive correlation was noted between the microdeletion and microinsertion frequencies for 564 genes for which both microdeletions and microinsertions are reported in HGMD, consistent with the view that the propensity of a given gene/sequence to undergo microdeletion is related to its propensity to undergo microinsertion. While microdeletions and microinsertions of 1 bp constitute respectively 48% and 66% of the corresponding totals, the relative frequency of the remaining lesions correlates negatively with the length of the DNA sequence deleted or inserted. Many of the microdeletions and microinsertions of more than 1 bp are potentially explicable in terms of slippage mutagenesis, involving the addition or removal of one copy of a mono-, di-, or trinucleotide tandem repeat. The frequency of in-frame 3-bp and 6-bp microinsertions and microdeletions was, however, found to be significantly lower than that of mutations of other lengths, suggesting that some of these in-frame lesions may not have come to clinical attention. Various sequence motifs were found to be over-represented in the vicinity of both microinsertions and microdeletions, including the heptanucleotide CCCCCTG that shares homology with the complement of the 8-bp human minisatellite conserved sequence/chi-like element (GCWGGWGG). The previously reported indel hotspot GTAAGT and its complement ACTTAC were also found to be overrepresented in the vicinity of both microinsertions and microdeletions, thereby providing a first example of a mutational hotspot that is common to different types of gene lesion. Other motifs overrepresented in the vicinity of microdeletions and microinsertions included DNA polymerase pause sites and topoisomerase cleavage sites. Several novel microdeletion/microinsertion hotspots were noted and some of these exhibited sufficient similarity to one another to justify terming them "super-hotspot" motifs. Analysis of sequence complexity also demonstrated that a combination of slipped mispairing mediated by direct repeats, and secondary structure formation promoted by symmetric elements, can account for the majority of microdeletions and microinsertions. Thus, microinsertions and microdeletions exhibit strong similarities in terms of the characteristics of their flanking DNA sequences, implying that they are generated by very similar underlying mechanisms.
Collapse
Affiliation(s)
- Edward V Ball
- Institute of Medical Genetics, Cardiff University, Cardiff, United Kingdom
| | | | | | | | | | | |
Collapse
|
22
|
Kim BG, Li C, Qiao W, Mamura M, Kasprzak B, Kasperczak B, Anver M, Wolfraim L, Hong S, Mushinski E, Potter M, Kim SJ, Fu XY, Deng C, Letterio JJ. Smad4 signalling in T cells is required for suppression of gastrointestinal cancer. Nature 2006; 441:1015-9. [PMID: 16791201 DOI: 10.1038/nature04846] [Citation(s) in RCA: 253] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2006] [Accepted: 04/28/2006] [Indexed: 11/08/2022]
Abstract
SMAD4 (MAD homologue 4 (Drosophila)), also known as DPC4 (deleted in pancreatic cancer), is a tumour suppressor gene that encodes a central mediator of transforming growth factor-beta signalling. Germline mutations in SMAD4 are found in over 50% of patients with familial juvenile polyposis, an autosomal dominant disorder characterized by predisposition to hamartomatous polyps and gastrointestinal cancer. Dense inflammatory cell infiltrates underlay grossly normal appearing, non-polypoid colonic and gastric mucosa of patients with familial juvenile polyposis. This prominent stromal component suggests that loss of SMAD4-dependent signalling in cells within the epithelial microenvironment has an important role in the evolution of intestinal tumorigenesis in this syndrome. Here we show that selective loss of Smad4-dependent signalling in T cells leads to spontaneous epithelial cancers throughout the gastrointestinal tract in mice, whereas epithelial-specific deletion of the Smad4 gene does not. Tumours arising within the colon, rectum, duodenum, stomach and oral cavity are stroma-rich with dense plasma cell infiltrates. Smad4(-/-) T cells produce abundant T(H)2-type cytokines including interleukin (IL)-5, IL-6 and IL-13, known mediators of plasma cell and stromal expansion. The results support the concept that cancer, as an outcome, reflects the loss of the normal communication between the cellular constituents of a given organ, and indicate that Smad4-deficient T cells ultimately send the wrong message to their stromal and epithelial neighbours.
Collapse
Affiliation(s)
- Byung-Gyu Kim
- Laboratory of Cell Regulation and Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
Juvenile polyposis syndrome (JPS) is an autosomal dominant cancer predisposition syndrome characterized by congenital anomalies, hamartomatous polyps in the gastrointestinal tract, and the development of tumors in these tissues. The diagnosis of JPS is often difficult because of the phenotypic overlap with other hamartomatous polyposis syndromes. Germline mutations have been identified in MADH4 and BMPR1A, aiding in presymptomatic genetic testing. In this study, we describe the results from 3 years of molecular diagnostic screening in JPS. Seventy unrelated individuals referred to our lab for JPS testing were examined through the sequence analysis of coding regions and exon-intron boundaries in both genes. Germline mutations were identified in 30% of cases, with 11.4% in BMPR1A and 18.6% in MADH4. All mutation-positive individuals were negative for cancer at testing, and a single pulmonary valve stenosis was the only congenital anomaly reported. A majority of mutations identified were novel including the first splice site alteration in MADH4. Based on the limited number of exons in each gene, low polymorphism frequency, and high frequency of frameshift or nonsense mutations identified, direct sequence analysis is a suitable methodology for mutation screening if all coding regions and exon-intron boundaries are examined in both genes.
Collapse
Affiliation(s)
- Robert E Pyatt
- Department of Pathology, Ohio State University, Hamilton Hall 125, 1645 Neil Ave., Columbus, OH 43210, USA
| | | | | |
Collapse
|
24
|
Abstract
Juvenile Polyposis Syndrome is an uncommon hamartomatous disorder with significant gastrointestinal malignant potential. Mutations in SMAD4 and BMPR1A, implicated in the Transforming Growth Factor beta pathway, have recently been characterized, and hold significance in the management of patients and at risk family members. This article reviews our knowledge to date of the genetics and clinicopathological features of the Juvenile Polyposis Syndrome, and discusses the current expert recommendations for genetic testing, disease screening and management.
Collapse
Affiliation(s)
- Elizabeth Chow
- Department of Colorectal Medicine and Genetics, Royal Melbourne Hospital, Victoria, Australia.
| | | |
Collapse
|
25
|
Affiliation(s)
- Anders Merg
- Roswell Park Cancer Institute, Buffalo, New York, USA
| | | | | | | |
Collapse
|
26
|
Shikata K, Kukita Y, Matsumoto T, Esaki M, Yao T, Mochizuki Y, Hayashi K, Iida M. Gastric juvenile polyposis associated with germline SMAD4 mutation. Am J Med Genet A 2005; 134:326-9. [PMID: 15754356 DOI: 10.1002/ajmg.a.30482] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We treated a 39-year-old woman with hypoproteinemia and anemia who had profuse gastric polyposis. Radiographic and endoscopic examination showed numerous polyps restricted to the stomach. The patient had pulmonary arteriovenous malformations in the left lung. Histological examination of the resected stomach revealed the gastric polyposis to be composed of cystic dilatation of the glands with small areas of adenocarcinoma. These findings were compatible with gastric juvenile polyposis (GJP) accompanied by gastric cancer. Analysis of genomic DNA revealed that the patient had truncating mutation of SMAD4, a responsible gene for juvenile polyposis (JP). Our case suggests that SMAD4 is possibly a responsible gene for GJP.
Collapse
Affiliation(s)
- Kentaro Shikata
- Department of Medicine and Clinical Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Howe JR, Sayed MG, Ahmed AF, Ringold J, Larsen-Haidle J, Merg A, Mitros FA, Vaccaro CA, Petersen GM, Giardiello FM, Tinley ST, Aaltonen LA, Lynch HT. The prevalence of MADH4 and BMPR1A mutations in juvenile polyposis and absence of BMPR2, BMPR1B, and ACVR1 mutations. J Med Genet 2004; 41:484-91. [PMID: 15235019 PMCID: PMC1735829 DOI: 10.1136/jmg.2004.018598] [Citation(s) in RCA: 194] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
BACKGROUND Juvenile polyposis (JP) is an autosomal dominant syndrome predisposing to colorectal and gastric cancer. We have identified mutations in two genes causing JP, MADH4 and bone morphogenetic protein receptor 1A (BMPR1A): both are involved in bone morphogenetic protein (BMP) mediated signalling and are members of the TGF-beta superfamily. This study determined the prevalence of mutations in MADH4 and BMPR1A, as well as three other BMP/activin pathway candidate genes in a large number of JP patients. METHODS DNA was extracted from the blood of JP patients and used for PCR amplification of each exon of these five genes, using primers flanking each intron-exon boundary. Mutations were determined by comparison to wild type sequences using sequence analysis software. A total of 77 JP cases were sequenced for mutations in the MADH4, BMPR1A, BMPR1B, BMPR2, and/or ACVR1 (activin A receptor) genes. The latter three genes were analysed when MADH4 and BMPR1A sequencing found no mutations. RESULTS Germline MADH4 mutations were found in 14 cases (18.2%) and BMPR1A mutations in 16 cases (20.8%). No mutations were found in BMPR1B, BMPR2, or ACVR1 in 32 MADH4 and BMPR1A mutation negative cases. DISCUSSION In the largest series of JP patients reported to date, the prevalence of germline MADH4 and BMPR1A mutations is approximately 20% for each gene. Since mutations were not found in more than half the JP patients, either additional genes predisposing to JP remain to be discovered, or alternate means of inactivation of the two known genes are responsible for these JP cases.
Collapse
Affiliation(s)
- J R Howe
- Department of Surgery, University of Iowa College of Medicine, Iowa City, IA 52242-1086, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Fogt F, Brown CA, Badizadegan K, Zimmerman RL, Odze R. Low prevalence of loss of heterozygosity and SMAD4 mutations in sporadic and familial juvenile polyposis syndrome-associated juvenile polyps. Am J Gastroenterol 2004; 99:2025-31. [PMID: 15447767 DOI: 10.1111/j.1572-0241.2004.30502.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Juvenile polyps (JP) may develop sporadically or may be associated with the familial juvenile polyposis syndrome (FJPS). In FJPS, the epithelium is susceptible to dysplasia and, ultimately adenocarcinoma. However, the mechanisms involved in this transformation are unknown. Since the epithelium in colorectal carcinogenesis undergoes a stepwise genetic progression, the purpose of this study was to determine if loss of heterozygosity (LOH) abnormalities can aid in the differentiation between sporadic and FJPS-associated polyps. DESIGN Ninety-one routinely-processed JP from three groups of patients were evaluated for this study. Group 1 included 39 polyps from 39 patients with a single JP and no personal or family history of FJPS; group 2 consisted of 24 polyps from 15 patients with 2-5 JP and no history of FJPS; and group 3 included 29 polyps from 22 patients with > or =5 polyps either with (7) or without (15) a family history of FJPS. Epithelium from typical, atypical, and overtly dysplastic polyps, when present (2 cases in group 3 only), were evaluated separately by microdissection and PCR analysis for LOH of APC, p53, 3p, 9p, and mutations in exon 9 of the SMAD4 gene. RESULTS SMAD4 mutations were observed in 3 polyps from 2 patients in group 3 (10% of informative cases; p < 0.05 vs group 1), but not in any of the polyps from the other two groups. Overall, LOH of APC, p53, 3p, and 9p were detected in 1%, 15%, 10%, and 4% of JPs, but no differences were observed between the three clinical groups. Two polyps, both in group 3, with definite dysplasia did not show any genetic alterations. The morphologic appearance of the polyps was not a reliable feature in helping to differentiate sporadic from FJPS-associated polyps. CONCLUSIONS LOH of APC, p53, 3p, and 9p may not be involved in the carcinogenic pathway of FJPS-associated polyps. SMAD4 gene mutations show a low sensitivity but a high specificity for FJPS.
Collapse
Affiliation(s)
- Franz Fogt
- Department of Pathology, University of Pennsylvania, Presbyterian Medical Center, 39th and Market Streets, Philadelphia, PA 19104, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
Juvenile polyps are hamartomatous polyps found primarily in infants and children, and in association with juvenile polyposis (JP; OMIM #174900), Cowden syndrome (CS; OMIM #158350), and Bannayan-Riley-Ruvalcaba syndrome (BRRS; OMIM# 153480). Although solitary juvenile polyps are benign lesions, when present in JP patients they may lead to gastrointestinal cancers. Germline mutations in MADH4 and BMPR1A predispose to JP, and both genes are involved in TGF-beta superfamily signaling pathways. In CS and BRRS, juvenile polyps are a less consistent feature, and CS patients are at risk for breast and thyroid cancers. Mutations of the tumor suppressor gene PTEN have been found in the germline of both CS and BRRS patients. Despite different underlying genetic mechanisms, these and other syndromes share the same phenotypic feature of juvenile polyps.
Collapse
Affiliation(s)
- Anders Merg
- Surgical Oncology and Endocrine Surgery, Department of Surgery, Roy J. and Lucille A. Carver Iowa College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | | |
Collapse
|
30
|
Waite KA, Eng C. From developmental disorder to heritable cancer: it's all in the BMP/TGF-beta family. Nat Rev Genet 2003; 4:763-73. [PMID: 14526373 DOI: 10.1038/nrg1178] [Citation(s) in RCA: 222] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Transforming growth factor-beta (TGF-beta) regulates many cellular processes through complex signal-transduction pathways that have crucial roles in normal development. Disruption of these pathways can lead to a range of diseases, including cancer. Mutations in the genes that encode members of the TGF-beta pathway are involved in vascular diseases as well as gastrointestinal neoplasia. More recently, they have been implicated in Cowden syndrome, which is normally associated with mutations in the phosphatase and tensin homologue gene PTEN. Molecular studies of TGF-beta signalling are now showing why mutations in genes that encode components of this pathway result in inherited cancer and developmental diseases.
Collapse
Affiliation(s)
- Kristin A Waite
- Human Cancer Genetics and Clinical Cancer Genetics Programs, Comprehensive Cancer Center, Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, Ohio 43210, USA
| | | |
Collapse
|
31
|
Kikkawa Y, Oyama A, Ishii R, Miura I, Amano T, Ishii Y, Yoshikawa Y, Masuya H, Wakana S, Shiroishi T, Taya C, Yonekawa H. A Small Deletion Hotspot in the Type II Keratin Gene mK6irs1/Krt2-6g on Mouse Chromosome 15, a Candidate for Causing the Wavy Hair of the Caracul (Ca) Mutation. Genetics 2003; 165:721-33. [PMID: 14573483 PMCID: PMC1462786 DOI: 10.1093/genetics/165.2.721] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Abstract
A new mutation has arisen in a colony of mice transgenic for human α-galactosidase. The mutation is independent of the transgenic insertion, autosomal dominant, and morphologically very similar to the classical wavy coat mutation, caracul (Ca), on chromosome 15. Therefore, we designated this locus the caracul Rinshoken (CaRin). Applying a positional cloning approach, we identified the mK6irs1/Krt2-6g gene as a strong candidate for CaRin because among five Ca alleles examined mutations always occurred in the highly conserved positions of the α-helical rod domain (1A and 2B subdomain) of this putative gene product. The most striking finding is that four independently discovered alleles, the three preexistent alleles CaJ, Ca9J, Ca10J, and our allele CaRin, all share one identical amino acid deletion (N 140 del) and the fifth, CamedJ, has an amino acid substitution (A 431 D). These findings indicate that a mutation hotspot exists in the Ca locus. Additionally, we describe a Ca mutant allele induced by ENU mutagenesis, which also possesses an amino acid substitution (L 424 W) in the mK6irs1/Krt2-6g gene. The identification of the Ca candidate gene enables us to further define the nature of the genetic pathway required for hair formation and provides an important new candidate that may be implicated in human hair and skin diseases.
Collapse
Affiliation(s)
- Yoshiaki Kikkawa
- Department of Laboratory Animal Science, The Tokyo Metropolitan Institute of Medical Science (Rinshoken), Tokyo 113-8613, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Colorectal cancer is the third leading cause of cancer-related deaths in both men and women in the United States and is estimated to have affected 148,000 people in 2002. The cumulative lifetime risk for colon cancer is approximately 5%-6%, and this risk is influenced by hereditary and lifestyle factors. In fact, 20%-30% of all colon cancer cases have a potentially definable inherited cause, and 3%-5% of colon cancers occur in genetically defined high-risk colon cancer family syndromes. Although the genes responsible for the cases of moderate-risk colon cancer remain to be characterized, many of the genes responsible for the high-risk colon cancer cases have already been determined. These genetic discoveries have been translated into clinical practice and have led to improved risk assessment through the use of genetic testing. The introduction into clinical practice of genetic testing for the assessment of colon cancer risk has led to more effective management strategies for patients with potentially high-risk colon cancer and has presented new challenges to the clinician because of the unique issues involved with genetic testing. In this review, an overview of the colon cancer high-risk syndromes, with a focus on the availability and indications for genetic testing, is presented.
Collapse
Affiliation(s)
- William M Grady
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37232-2279, USA.
| |
Collapse
|
33
|
Nagasako T, Sugiyama T, Mizushima T, Miura Y, Kato M, Asaka M. Up-regulated Smad5 mediates apoptosis of gastric epithelial cells induced by Helicobacter pylori infection. J Biol Chem 2003; 278:4821-5. [PMID: 12473652 DOI: 10.1074/jbc.m211143200] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The gastric pathogen Helicobacter pylori activates epithelial cell signaling pathways, and its infection induces changes in the expression of several genes in infected human gastric tissues. Recent studies have indicated that the ability of H. pylori to regulate epithelial cell responses depends on the presence of an intact cag pathogenicity island (cagPAI). We investigated altered mRNA expression of gastric epithelial cells after infection with H. pylori, both cagPAI-positive and cagPAI-negative strains, by cDNA microarray, reverse transcription PCR, and Northern blot analysis. Our results indicated that cagPAI-positive H. pylori strains (ATCC 43504 and clinical isolated strains) significantly activated Smad5 mRNA expression of human gastric epithelial cells (AGS, KATOIII, MKN28, and MKN45). We further examined whether the up-regulated Smad5 was related to apoptosis of gastric epithelial cells induced by H. pylori. Smad5 RNA interference completely inhibited H. pylori-induced apoptosis. These results suggest that Smad5 is up-regulated in gastric epithelial cells through the presence of cagPAI of H. pylori and that Smad5 mediates apoptosis of gastric epithelial cells induced by H. pylori infection.
Collapse
Affiliation(s)
- Tomokazu Nagasako
- Department of Gastroenterology, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan
| | | | | | | | | | | |
Collapse
|