1
|
Garcia-Solorio J, Núñez-Enriquez JC, Jiménez-Olivares M, Flores-Lujano J, Flores-Espino F, Molina-Garay C, Cervera A, Casique-Aguirre D, Peñaloza-Gonzalez JG, Baños-Lara MDR, García-Soto Á, Galván-Díaz CA, Olaya-Vargas A, Aguilar HF, Mata-Rocha M, Garrido-Hernández MÁ, Solís-Poblano JC, Luna-Silva NC, Cano-Cuapio LS, Aristil-Chery PM, Herrera-Quezada F, Carrillo-Sanchez K, Muñoz-Rivas A, Flores-Lagunes LL, Mendoza-Caamal EC, Villegas-Torres BE, González-Osnaya V, Jiménez-Hernández E, Torres-Nava JR, Martín-Trejo JA, Gutiérrez-Rivera MDL, Espinosa-Elizondo RM, Merino-Pasaye LE, Pérez-Saldívar ML, Jiménez-Morales S, Curiel-Quesada E, Rosas-Vargas H, Mejía-Arangure JM, Alaez-Verson C. IKZF1plus is a frequent biomarker of adverse prognosis in Mexican pediatric patients with B-acute lymphoblastic leukemia. Front Oncol 2024; 14:1337954. [PMID: 38634053 PMCID: PMC11022689 DOI: 10.3389/fonc.2024.1337954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 02/29/2024] [Indexed: 04/19/2024] Open
Abstract
Background Recurrent genetic alterations contributing to leukemogenesis have been identified in pediatric B-cell Acute Lymphoblastic Leukemia (B-ALL), and some are useful for refining classification, prognosis, and treatment selection. IKZF1plus is a complex biomarker associated with a poor prognosis. It is characterized by IKZF1 deletion coexisting with PAX5, CDKN2A/2B, or PAR1 region deletions. The mutational spectrum and clinical impact of these alterations have scarcely been explored in Mexican pediatric patients with B-ALL. Here, we report the frequency of the IKZF1plus profile and the mutational spectrum of IKZF1, PAX5, CDKN2A/2B, and ERG genes and evaluate their impact on overall survival (OS) in a group of patients with B-ALL. Methods A total of 206 pediatric patients with de novo B-ALL were included. DNA was obtained from bone marrow samples at diagnosis before treatment initiation. A custom-designed next-generation sequencing panel was used for mutational analysis. Kaplan-Meier analysis was used for OS estimation. Results We identified the IKZF1plus profile in 21.8% of patients, which was higher than that previously reported in other studies. A significantly older age (p=0.04), a trend toward high-risk stratification (p=0.06), and a decrease in 5-year Overall Survival (OS) (p=0.009) were observed, although heterogeneous treatment protocols in our cohort would have impacted OS. A mutation frequency higher than that reported was found for IKZF1 (35.9%) and CDKN2A/2B (35.9%) but lower for PAX5 (26.6%). IKZF1MUT group was older at diagnosis (p=0.0002), and most of them were classified as high-risk (73.8%, p=0.02), while patients with CDKN2A/2BMUT had a higher leukocyte count (p=0.01) and a tendency toward a higher percentage of blasts (98.6%, >50% blasts, p=0.05) than the non-mutated patients. A decrease in OS was found in IKZF1MUT and CDKN2A/2BMUT patients, but the significance was lost after IKZF1plus was removed. Discussion Our findings demonstrated that Mexican patients with B-ALL have a higher prevalence of genetic markers associated with poor outcomes. Incorporating genomic methodologies into the diagnostic process, a significant unmet need in low- and mid-income countries, will allow a comprehensive identification of relevant alterations, improving disease classification, treatment selection, and the general outcome.
Collapse
Affiliation(s)
- Joaquin Garcia-Solorio
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Juan Carlos Núñez-Enriquez
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Marco Jiménez-Olivares
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Janet Flores-Lujano
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Fernanda Flores-Espino
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Carolina Molina-Garay
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Alejandra Cervera
- Subdirección de Genómica Poblacional, Instituto Nacional de Medicina Genomica (INMEGEN), Mexico City, Mexico
| | - Diana Casique-Aguirre
- Laboratorio de Citómica del Cáncer Infantil, Centro de Investigación Biomédica de Oriente, Instituto Mexicano del Seguro Social, Delegación Puebla, Puebla, Mexico
- Consejo Nacional de Humanidades, Ciencias y Tecnologías (CONAHCYT), Mexico City, Mexico
| | | | - Ma. Del Rocío Baños-Lara
- Centro de Investigación Oncológica Una Nueva Esperanza, Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico
| | - Ángel García-Soto
- Hospital General Centro Médico La Raza, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Alberto Olaya-Vargas
- Departamento de Oncologia, Instituto Nacional de Pediatría (INP), Mexico City, Mexico
| | - Hilario Flores Aguilar
- Departamento de Inmunogenetica, Instituto de Diagnostico y Referencia Epidemiologicos (InDRE), Mexico City, Mexico
| | - Minerva Mata-Rocha
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | | | - Juan Carlos Solís-Poblano
- Servicio de Oncohematología Pediátrica, Instituto Mexicano del Seguro (IMSS) Unidad Médica de Alta Especialidad (UMAE) Centro Médico Nacional (CMN) Hospital de Especialidades Dr. Manuel Ávila Camacho, Puebla, Mexico
| | - Nuria Citlalli Luna-Silva
- Servicio de Hemato-Oncología Pediátrica, Hospital de la Niñez Oaxaqueña "Dr. Guillermo Zárate Mijangos", Secretaria de Salud y Servicios de Salud Oaxaca (SSO), Oaxaca, Mexico
| | | | - Pierre Mitchel Aristil-Chery
- Instituto de Seguridad y Servicios Sociales de los Trabajadores al Servicio de los Poderes del Estado (ISSSTE) de Puebla, Departamento de Enseñanza e Investigació, Puebla, Mexico
| | - Fernando Herrera-Quezada
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Karol Carrillo-Sanchez
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Anallely Muñoz-Rivas
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | | | | | | | - Vincent González-Osnaya
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Elva Jiménez-Hernández
- Servicio de Oncología, Hospital Pediátrico Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - José Refugio Torres-Nava
- Servicio de Oncología, Hospital Pediátrico Moctezuma, Secretaría de Salud de la Ciudad de México (SSCDMX), Mexico City, Mexico
| | - Jorge Alfonso Martín-Trejo
- Servicio de Hematología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría “Dr. Silvestre Frenk Freund”, Mexico City, Mexico
| | - María de Lourdes Gutiérrez-Rivera
- Servicio de Oncología, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Unidad Médica de Alta Especialidad (UMAE) Hospital de Pediatría “Dr. Silvestre Frenk Freund”, Mexico City, Mexico
| | | | - Laura Elizabeth Merino-Pasaye
- Servicio de Hematología Pediátrica, Centro Médico Nacional (CMN) “20 de Noviembre”, Instituto de Seguridad Social al Servicio de los Trabajadores del Estado (ISSSTE), Mexico City, Mexico
| | - María Luisa Pérez-Saldívar
- Unidad de Investigación Médica en Epidemiología Clínica, Unidad Medica de Alta Especialidad (UMAE) Hospital de Pediatría, Centro Médico Nacional (CMN) Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Silvia Jiménez-Morales
- Laboratorio de Medicina de Precisión, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Everardo Curiel-Quesada
- Departamento de Bioquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politecnico Nacional (IPN), Mexico City, Mexico
| | - Haydeé Rosas-Vargas
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, CMN Siglo XXI, Instituto Mexicano del Seguro Social (IMSS), Mexico City, Mexico
| | - Juan Manuel Mejía-Arangure
- Laboratorio de Genómica Funcional del Cáncer, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Facultad de Medicina, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
| | - Carmen Alaez-Verson
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| |
Collapse
|
2
|
Jeong AR, Forbes K, Orosco RK, Cohen EEW. Hereditary oral squamous cell carcinoma associated with CDKN2A germline mutation: a case report. J Otolaryngol Head Neck Surg 2022; 51:5. [PMID: 35123577 PMCID: PMC8818223 DOI: 10.1186/s40463-022-00556-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Accepted: 01/02/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Germline CDKN2A mutations are a well-known cause of familial atypical multiple mole melanoma (OMIM #155601) and melanoma-pancreatic cancer syndrome (OMIM #606719). Increased risk of head and neck squamous cell carcinoma (HNSCC), particularly oral squamous cell carcinoma (OSCC) in those with germline CDKN2A mutations, has been described. However, screening for HNSCC is not a routine practice in patients with CDKN2A germline mutations and these mutations are not a conventional test for HNSCC patients without obvious risk factors. CASE PRESENTATION We describe a female with no smoking history who developed oral squamous cell carcinoma at age 39 and had a complex clinical course of recurrent multifocal squamous cell carcinoma (SCC) and carcinoma in situ of the oral cavity and oropharynx. Detailed family history demonstrated that her mother was diagnosed with OSCC and melanoma in her 40 s, and her maternal grandfather was diagnosed with metastatic melanoma in his 40 s. Genetic testing of the patient and her mother revealed CDKN2A c.301G>T mutation. She was referred to genetic counseling as well as to dermatology, gastroenterology, and neurology for cancer surveillance. She was treated with resections and has no evidence of disease 3 years after diagnosis. CONCLUSIONS We report a family with a CDKN2A c.301 G>T mutation who also have significant history of OSCC, adding to the growing body of literature suggesting increased risk of HNSCC, particularly OSCC, in CDKN2A germline mutation carriers. It is important to consider CDKN2A mutation testing in familial HNSCC and young patients without obvious risk factors. Moreover, surveillance for HNSCC should be routine practice in those with a CDKN2A germline mutation.
Collapse
Affiliation(s)
- Ah-Reum Jeong
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0960, USA
| | - Kimberly Forbes
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0960, USA
| | - Ryan K Orosco
- Division of Otolaryngology, Department of Surgery, University of California San Diego, La Jolla, CA, 92093, USA.,Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ezra E W Cohen
- Division of Hematology and Oncology, Department of Medicine, University of California San Diego, 3855 Health Sciences Drive, La Jolla, CA, 92093-0960, USA. .,Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
3
|
Li W, Ma Z, Fu X, Hao Z, Shang H, Shi J, Lei M, Xu M, Ning S, Hua X. Olaparib effectively treats local recurrence of extrahepatic cholangiocarcinoma in a patient harboring a BRCA2-inactivating mutation: a case report. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1487. [PMID: 34734039 PMCID: PMC8506765 DOI: 10.21037/atm-21-3681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 09/02/2021] [Indexed: 12/31/2022]
Abstract
Cholangiocarcinoma (CCA) is a malignant tumor with poor prognosis and high recurrence rate. There is no standard treatment for advanced CCA beyond first-line chemotherapy, which provides only limited benefits. In this study, we report a case of a postoperative recurrence ECC patient harboring a breast cancer 2 (BRCA2)-inactivating rearrangement mutation that had an obvious reaction to olaparib therapy. The patient was a 68-year-old man with postoperative recurrence of extrahepatic CCA (ECC) who declined systemic chemotherapy. In August 2015, abdominal computed tomography (CT) of the patient revealed intrahepatic bile duct dilatation, obstruction at the hepatic hilar region proximal to the common hepatic duct, and splenomegaly, and radical surgical resection was performed. Postoperative histopathology diagnosis was ECC without metastases. In February 2017, abdominal CT revealed local recurrence, and the patient refused chemotherapy. BRCA2 rearrangement were detected by next-generation sequencing. Oral administration of olaparib was initiated. The patient achieved stable disease 1 month later, progression-free survival for >10 months without any significant adverse reactions, and an overall survival (OS) of 27 months. This is the first report demonstrating the clinical benefits of olaparib in a BRCA2 rearrangement-harboring patient with ECC. This observation would help determine the best treatment option for advanced ECC patients.
Collapse
Affiliation(s)
- Wenxin Li
- Department of Hepatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Zuohong Ma
- Department of Hepatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Xibo Fu
- Department of Hepatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Zhiqiang Hao
- Department of Hepatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Hai Shang
- Department of Hepatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| | | | | | - Mian Xu
- Shanghai OrigiMed Co. Ltd., Shanghai, China
| | - Shili Ning
- Department of General Surgery, The Second Hospital of Dalian Medical University, Dalian, China
| | - Xiangdong Hua
- Department of Hepatobiliary Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
4
|
CDKN2A-Mutated Pancreatic Ductal Organoids from Induced Pluripotent Stem Cells to Model a Cancer Predisposition Syndrome. Cancers (Basel) 2021; 13:cancers13205139. [PMID: 34680288 PMCID: PMC8533699 DOI: 10.3390/cancers13205139] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 10/05/2021] [Accepted: 10/09/2021] [Indexed: 12/20/2022] Open
Abstract
Patient-derived induced pluripotent stem cells (iPSCs) provide a unique platform to study hereditary disorders and predisposition syndromes by resembling germline mutations of affected individuals and by their potential to differentiate into nearly every cell type of the human body. We employed plucked human hair from two siblings with a family history of cancer carrying a pathogenic CDKN2A variant, P16-p.G101W/P14-p.R115L, to generate patient-specific iPSCs in a cancer-prone ancestry for downstream analytics. The differentiation capacity to pancreatic progenitors and to pancreatic duct-like organoids (PDLOs) according to a recently developed protocol remained unaffected. Upon inducible expression of KRASG12Dusing a piggyBac transposon system in CDKN2A-mutated PDLOs, we revealed structural and molecular changes in vitro, including disturbed polarity and epithelial-to-mesenchymal (EMT) transition. CDKN2A-mutated KRASG12DPDLO xenotransplants formed either a high-grade precancer lesion or a partially dedifferentiated PDAC-like tumor. Intriguingly, P14/P53/P21 and P16/RB cell-cycle checkpoint controls have been only partly overcome in these grafts, thereby still restricting the tumorous growth. Hereby, we provide a model for hereditary human pancreatic cancer that enables dissection of tumor initiation and early development starting from patient-specific CDKN2A-mutated pluripotent stem cells.
Collapse
|
5
|
Yamashita T, Vollbrecht C, Hirsch B, Kleo K, Anagnostopoulos I, Hummel M. Integrative genomic analysis focused on cell cycle genes for MYC-driven aggressive mature B-cell lymphoma. J Clin Exp Hematop 2021; 60:87-96. [PMID: 32981916 PMCID: PMC7596913 DOI: 10.3960/jslrt.20021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
MYC is a transcriptional factor that regulates growth and proliferation through cell
cycle pathways. MYC alterations, in particular MYC rearrangements, are
important in assessing the prognosis of aggressive B-cell lymphoma. In this study, we
focused on the impact of nine major cell cycle genes for MYC-driven aggressive mature
B-cell lymphoma and analyzed the mutational status using targeted next generation
sequencing. Our 40 cases of aggressive mature B-cell lymphomas included 5 Burkitt
lymphomas, 17 high-grade B-cell lymphomas and 18 diffuse large B-cell lymphomas with MYC
breaks in 100%, 88% and 11%, respectively. Our data allowed a molecular classification
into four categories partially independent from the histopathological diagnosis but
correlating with the Ki-67 labelling index: (I) harboring TP53 and
CDKN2A mutations, being highly proliferative, (II) with
MYC rearrangement associated with MYC and/or
ID3 mutations, being highly proliferative, (III) with
MYC rearrangement combined with additional molecular changes, being
highly proliferative, and (IV) with a diverse pattern of molecular alterations, being less
proliferative. Taken together, we found that mutations of TP53,
CDKN2A, MYC and ID3 are associated
with highly proliferative B-cell lymphomas that could profit from novel therapeutic
strategies.
Collapse
Affiliation(s)
- Takahisa Yamashita
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,Department of Pathology, Saitama Medical Center, Saitama Medical University, Saitama, Japan
| | - Claudia Vollbrecht
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany.,German Cancer Consortium (DKTK), Berlin, Germany.,German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Burkhard Hirsch
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Karsten Kleo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Ioannis Anagnostopoulos
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| | - Michael Hummel
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Institute of Pathology, Berlin, Germany
| |
Collapse
|
6
|
Genetic Alterations in the INK4a/ARF Locus: Effects on Melanoma Development and Progression. Biomolecules 2020; 10:biom10101447. [PMID: 33076392 PMCID: PMC7602651 DOI: 10.3390/biom10101447] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 10/11/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023] Open
Abstract
Genetic alterations in the INK4a/ARF (or CDKN2A) locus have been reported in many cancer types, including melanoma; head and neck squamous cell carcinomas; lung, breast, and pancreatic cancers. In melanoma, loss of function CDKN2A alterations have been identified in approximately 50% of primary melanomas, in over 75% of metastatic melanomas, and in the germline of 40% of families with a predisposition to cutaneous melanoma. The CDKN2A locus encodes two critical tumor suppressor proteins, the cyclin-dependent kinase inhibitor p16INK4a and the p53 regulator p14ARF. The majority of CDKN2A alterations in melanoma selectively target p16INK4a or affect the coding sequence of both p16INK4a and p14ARF. There is also a subset of less common somatic and germline INK4a/ARF alterations that affect p14ARF, while not altering the syntenic p16INK4a coding regions. In this review, we describe the frequency and types of somatic alterations affecting the CDKN2A locus in melanoma and germline CDKN2A alterations in familial melanoma, and their functional consequences in melanoma development. We discuss the clinical implications of CDKN2A inactivating alterations and their influence on treatment response and resistance.
Collapse
|
7
|
Liu Y, Li C, Shen S, Chen X, Szlachta K, Edmonson MN, Shao Y, Ma X, Hyle J, Wright S, Ju B, Rusch MC, Liu Y, Li B, Macias M, Tian L, Easton J, Qian M, Yang JJ, Hu S, Look AT, Zhang J. Discovery of regulatory noncoding variants in individual cancer genomes by using cis-X. Nat Genet 2020; 52:811-818. [PMID: 32632335 PMCID: PMC7679232 DOI: 10.1038/s41588-020-0659-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 06/05/2020] [Indexed: 12/30/2022]
Abstract
We developed cis-X, a computational method for discovering regulatory noncoding variants in cancer by integrating whole-genome and transcriptome sequencing data from a single cancer sample. cis-X first finds aberrantly cis-activated genes that exhibit allele-specific expression accompanied by an elevated outlier expression. It then searches for causal noncoding variants that may introduce aberrant transcription factor binding motifs or enhancer hijacking by structural variations. Analysis of 13 T-lineage acute lymphoblastic leukemias identified a recurrent intronic variant predicted to cis-activate the TAL1 oncogene, a finding validated in vivo by chromatin immunoprecipitation sequencing of a patient-derived xenograft. Candidate oncogenes include the prolactin receptor PRLR activated by a focal deletion that removes a CTCF-insulated neighborhood boundary. cis-X may be applied to pediatric and adult solid tumors that are aneuploid and heterogeneous. In contrast to existing approaches, which require large sample cohorts, cis-X enables the discovery of regulatory noncoding variants in individual cancer genomes.
Collapse
Affiliation(s)
- Yu Liu
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China. .,Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Chunliang Li
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shuhong Shen
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaolong Chen
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Karol Szlachta
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael N Edmonson
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Ying Shao
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Xiaotu Ma
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Judith Hyle
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shaela Wright
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Bensheng Ju
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Michael C Rusch
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Yanling Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Benshang Li
- Pediatric Translational Medicine Institute, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Key Laboratory of Pediatric Hematology & Oncology Ministry of Health, Department of Hematology & Oncology, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Michael Macias
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Liqing Tian
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - John Easton
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maoxiang Qian
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Jun J Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, TN, USA.,Hematological Malignancies Program, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shaoyan Hu
- Children's Hospital of Soochow University, Suzhou, China
| | - A Thomas Look
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Division of Pediatric Hematology-Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
8
|
Guyot A, Duchesne M, Robert S, Lia AS, Derouault P, Scaon E, Lemnos L, Salle H, Durand K, Labrousse F. Analysis of CDKN2A gene alterations in recurrent and non-recurrent meningioma. J Neurooncol 2019; 145:449-459. [PMID: 31729637 DOI: 10.1007/s11060-019-03333-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 11/03/2019] [Indexed: 12/11/2022]
Abstract
PURPOSE Assessment of the risk of recurrence is essential to determine the therapeutic strategy of meningioma treatment. Many relapsing or aggressive meningiomas show elevated mitotic and/or Ki67 indices, reflecting cell cycle deregulation. As CDKN2A is a key tumor suppressor gene involved in cell cycle control, we investigated whether CDKN2A alterations may be involved in tumor recurrence. METHODS We carried out a comparative analysis of 17 recurrent and 13 non-recurrent meningiomas. CDKN2A single nucleotide variations (SNVs), deletions, methylation status of the promotor, and p16 expression were investigated. Results were correlated with the recurrent or non-recurrent status and clinicopathological data. RESULTS We identified a CDKN2A SNV (NM_000077, exon2, c.G442A, p.Ala148Thr) in five meningiomas that was significantly associated with recurrence (p = 0.03). This mutation, confirmed by Sanger sequencing and referenced in the COSMIC database in various cancers, has not been reported in meningioma. The presence of one of the three following CDKN2A alterations-p.(Ala148Thr) mutation, whole homozygous or heterozygous gene loss, or promotor methylation > 8%-was observed in 13 of the 17 relapsing meningiomas and was strongly associated with recurrence (p < 0.0001) and a Ki67 labeling index > 7% (p = 0.004). CONCLUSION We report an undescribed p.(Ala148Thr) CDKN2A mutation in meningioma that was only present in relapsing tumors. In our series, CDKN2A gene alterations were only found in recurrent meningiomas. However, our results need to be evaluated on a larger series to ensure that these CDKN2A alterations can be used as biomarkers of recurrence in meningioma.
Collapse
Affiliation(s)
- Anne Guyot
- Department of Pathology, Limoges University Hospital, 2 Avenue Martin-Luther-King, 87042, Limoges, France
| | - Mathilde Duchesne
- Department of Pathology, Limoges University Hospital, 2 Avenue Martin-Luther-King, 87042, Limoges, France
| | - Sandrine Robert
- EA 3842, CAPTuR « Contrôle de L'Activation Cellulaire, Progression Tumorale Et Résistance Thérapeutique », Faculty of Medicine, Limoges University, 2 Rue du Docteur Marcland, 87025, Limoges, France
| | - Anne-Sophie Lia
- EA 6309, MMNP « Maintenance Myélinique Et Neuropathies Périphériques », Faculty of Medicine, Limoges University, 2 Rue du Docteur Marcland, 87025, Limoges, France
| | - Paco Derouault
- EA 6309, MMNP « Maintenance Myélinique Et Neuropathies Périphériques », Faculty of Medicine, Limoges University, 2 Rue du Docteur Marcland, 87025, Limoges, France
| | - Erwan Scaon
- Bioinformatics Unit, BISCEM Platform, CBRS, University of Limoges, 2 Rue du Docteur-Marcland, 87025, Limoges, France
| | - Leslie Lemnos
- Department of Neurosurgery, Limoges University Hospital, 2 Avenue Martin-Luther-King, 87042, Limoges, France
| | - Henri Salle
- Department of Neurosurgery, Limoges University Hospital, 2 Avenue Martin-Luther-King, 87042, Limoges, France
| | - Karine Durand
- Department of Pathology, Limoges University Hospital, 2 Avenue Martin-Luther-King, 87042, Limoges, France.,EA 3842, CAPTuR « Contrôle de L'Activation Cellulaire, Progression Tumorale Et Résistance Thérapeutique », Faculty of Medicine, Limoges University, 2 Rue du Docteur Marcland, 87025, Limoges, France
| | - François Labrousse
- Department of Pathology, Limoges University Hospital, 2 Avenue Martin-Luther-King, 87042, Limoges, France. .,EA 3842, CAPTuR « Contrôle de L'Activation Cellulaire, Progression Tumorale Et Résistance Thérapeutique », Faculty of Medicine, Limoges University, 2 Rue du Docteur Marcland, 87025, Limoges, France.
| |
Collapse
|
9
|
Alam MNU. Computational assessment of somatic and germline mutations of p16INK4a: Structural insights and implications in disease. INFORMATICS IN MEDICINE UNLOCKED 2019. [DOI: 10.1016/j.imu.2019.100208] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
|
10
|
Han GC, Hwang J, Wankowicz SAM, Zhang Z, Liu D, Cibulskis C, Gaviola GC, Ghazikhanian V, McKay RR, Bubley GJ, Carter SL, Balk SP, Hahn WC, Taplin ME, Van Allen EM. Genomic Resistance Patterns to Second-Generation Androgen Blockade in Paired Tumor Biopsies of Metastatic Castration-Resistant Prostate Cancer. JCO Precis Oncol 2017; 1:1700140. [PMID: 32913968 PMCID: PMC7446377 DOI: 10.1200/po.17.00140] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Patients with castration-resistant prostate cancer (CRPC) receive second-generation androgen-deprivation therapy, but frequently experience relapse or do not respond. Understanding the genetic mechanisms of resistance will help to identify strategies and biomarkers that are essential for the next line of therapy. Patients and Methods We analyzed whole exomes of patient-matched pre- and post-treatment tumors from patients with CRPC. These patients had received the secondary androgen-deprivation therapy agent, abiraterone, which suppresses androgens to below castration levels, or enzalutamide, which competitively inhibits the key androgen signaling effector, androgen receptor. Results We observed that abiraterone-resistant tumors harbored alterations in AR and MYC, whereas enzalutamide-resistant tumors gained alterations in cell-cycle pathway genes, such as mutation in cyclin-dependent kinase N2A (CDKN2A) or amplification of CDK6. Experimentally, overexpressing cell-cycle kinases promoted enzalutamide resistance in androgen-sensitive LnCAP cells that was mitigated via CDK4/6 blockade—palbociclib and ribociclib. Conclusion CDK4/6-mediated resistance observed in preclinical experiments suggests that CDK4/6 amplifications may sufficiently promote enzalutamide resistance in CRPC, and that these patients may respond to palbociclib or ribociclib. The overall observations suggest that, in genomically selected advanced CRPC, clinical strategies against abiraterone- or enzalutamide-resistant tumors may require treatment strategies that are tailored to the resistance mechanisms that are specific to those patient subpopulations.
Collapse
Affiliation(s)
- G Celine Han
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Justin Hwang
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Stephanie A M Wankowicz
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Zhenwei Zhang
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - David Liu
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Carrie Cibulskis
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Glenn C Gaviola
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Varand Ghazikhanian
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Rana R McKay
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Glenn J Bubley
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Scott L Carter
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Steven P Balk
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - William C Hahn
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Mary-Ellen Taplin
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| | - Eliezer M Van Allen
- , , , , , , , , and , Dana-Farber Cancer Institute; , , , and , Brigham and Women's Hospital; and , Beth Israel Deaconess Medical Center, Boston; , , , , , , , and , Broad Institute of Harvard and Massachusetts Institute of Technology, Cambridge, MA; and , University of California San Diego, La Jolla, CA
| |
Collapse
|
11
|
Koulermou G, Shammas C, Vassiliou A, Kyriakides TC, Costi C, Neocleous V, Phylactou LA, Pantelidou M. CDKN2A and MC1R variants found in Cypriot patients diagnosed with cutaneous melanoma. J Genet 2017; 96:155-160. [PMID: 28360400 DOI: 10.1007/s12041-017-0742-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The prevalence of genetic variants associated to cutaneous melanoma (CM) has never been determined within Cypriot melanomas. This study evaluates the frequency of variants in cyclin-dependent kinase inhibitor 2A (CDKN2A) and melanocortin-1 receptor (MC1R) in 32 patients diagnosed with CM. Other characteristics and risk factors were also assessed. CDKN2A p.Ala148Thr was detected in three of 32 patients, while the control group revealed no variations within CDKN2A. MC1R screening in 32 patients revealed the following variations: p.Val60Leu in 11 patients, p.Arg142His in four patients, p.Thr314Thr in one patient, p.Arg160Trp in one patient, p.Val92Met/p.Thr314Thr in one patient and p.Val92Met/p.Arg142His/p.Thr314Thr in one patient. The control group revealed only p.Val60Leu (in 10 of 45 individuals), which is frequently found in general populations. Two unrelated patients carried CDKN2A p.Ala148Thr in combination with MC1R p.Arg142His, suggesting digenic inheritance that may provide evidence of different gene variants acting synergistically to contribute for CM development. This study confirms the presence of CDKN2A and MC1R variants among Cypriot melanomas and supports existing evidence of a role for these variants in susceptibility to melanoma.
Collapse
Affiliation(s)
- Georgia Koulermou
- Department of Plastic Surgery and Burn Unit, Nicosia General Hospital, Nicosia 2029, Cyprus.
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Walsh KM, de Smith AJ, Hansen HM, Smirnov IV, Gonseth S, Endicott AA, Xiao J, Rice T, Fu CH, McCoy LS, Lachance DH, Eckel-Passow JE, Wiencke JK, Jenkins RB, Wrensch MR, Ma X, Metayer C, Wiemels JL. A Heritable Missense Polymorphism in CDKN2A Confers Strong Risk of Childhood Acute Lymphoblastic Leukemia and Is Preferentially Selected during Clonal Evolution. Cancer Res 2015; 75:4884-94. [PMID: 26527286 PMCID: PMC4651745 DOI: 10.1158/0008-5472.can-15-1105] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2015] [Accepted: 08/26/2015] [Indexed: 01/07/2023]
Abstract
Genome-wide association studies (GWAS) have identified SNPs in six genes that are associated with childhood acute lymphoblastic leukemia (ALL). A lead SNP was found to occur on chromosome 9p21.3, a region that is deleted in 30% of childhood ALLs, suggesting the presence of causal polymorphisms linked to ALL risk. We used SNP genotyping and imputation-based fine-mapping of a multiethnic ALL case-control population (Ncases = 1,464, Ncontrols = 3,279) to identify variants of large effect within 9p21.3. We identified a CDKN2A missense variant (rs3731249) with 2% allele frequency in controls that confers three-fold increased risk of ALL in children of European ancestry (OR, 2.99; P = 1.51 × 10(-9)) and Hispanic children (OR, 2.77; P = 3.78 × 10(-4)). Moreover, of 17 patients whose tumors displayed allelic imbalance at CDKN2A, 14 preferentially retained the risk allele and lost the protective allele (PBinomial = 0.006), suggesting that the risk allele provides a selective advantage during tumor growth. Notably, the CDKN2A variant was not significantly associated with melanoma, glioblastoma, or pancreatic cancer risk, implying that this polymorphism specifically confers ALL risk but not general cancer risk. Taken together, our findings demonstrate that coding polymorphisms of large effect can underlie GWAS "hits" and that inherited polymorphisms may undergo directional selection during clonal expansion of tumors.
Collapse
Affiliation(s)
- Kyle M Walsh
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California.
| | - Adam J de Smith
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Helen M Hansen
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Ivan V Smirnov
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Semira Gonseth
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Alyson A Endicott
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Jianqiao Xiao
- Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California
| | - Terri Rice
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Cecilia H Fu
- Division of Hematology/Oncology, Children's Hospital Los Angeles, Los Angeles, California
| | - Lucie S McCoy
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California
| | - Daniel H Lachance
- Department of Neurology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Jeanette E Eckel-Passow
- Division of Biomedical Statistics and Informatics, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - John K Wiencke
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Robert B Jenkins
- Department of Laboratory Medicine and Pathology, Mayo Clinic College of Medicine, Rochester, Minnesota
| | - Margaret R Wrensch
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| | - Xiaomei Ma
- Department of Chronic Disease Epidemiology, Yale University School of Public Health, New Haven, Connecticut
| | - Catherine Metayer
- School of Public Health, University of California Berkeley, Berkeley, California
| | - Joseph L Wiemels
- Division of Neuroepidemiology, Department of Neurological Surgery, University of California, San Francisco, San Francisco, California. Department of Epidemiology and Biostatistics, University of California, San Francisco, San Francisco, California. Institute for Human Genetics, University of California, San Francisco, San Francisco, California
| |
Collapse
|
13
|
Vijayakrishnan J, Henrion M, Moorman AV, Fiege B, Kumar R, Inacio da Silva Filho M, Holroyd A, Koehler R, Thomsen H, Irving JA, Allan JM, Lightfoot T, Roman E, Kinsey SE, Sheridan E, Thompson PD, Hoffmann P, Nöthen MM, Mühleisen TW, Eisele L, Bartram CR, Schrappe M, Greaves M, Hemminki K, Harrison CJ, Stanulla M, Houlston RS. The 9p21.3 risk of childhood acute lymphoblastic leukaemia is explained by a rare high-impact variant in CDKN2A. Sci Rep 2015; 5:15065. [PMID: 26463672 PMCID: PMC4604478 DOI: 10.1038/srep15065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 08/12/2015] [Indexed: 02/08/2023] Open
Abstract
Genome-wide association studies (GWAS) have provided strong evidence for inherited predisposition to childhood acute lymphoblastic leukaemia (ALL) identifying a number of risk loci. We have previously shown common SNPs at 9p21.3 influence ALL risk. These SNP associations are generally not themselves candidates for causality, but simply act as markers for functional variants. By means of imputation of GWAS data and subsequent validation SNP genotyping totalling 2,177 ALL cases and 8,240 controls, we have shown that the 9p21.3 association can be ascribed to the rare high-impact CDKN2A p.Ala148Thr variant (rs3731249; Odds ratio = 2.42, P = 3.45 × 10(-19)). The association between rs3731249 genotype and risk was not specific to particular subtype of B-cell ALL. The rs3731249 variant is associated with predominant nuclear localisation of the CDKN2A transcript suggesting the functional effect of p.Ala148Thr on ALL risk may be through compromised ability to inhibit cyclin D within the cytoplasm.
Collapse
Affiliation(s)
- Jayaram Vijayakrishnan
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Marc Henrion
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Anthony V. Moorman
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Bettina Fiege
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | | | - Amy Holroyd
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Rolf Koehler
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Hauke Thomsen
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
| | - Julie A. Irving
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - James M. Allan
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Tracy Lightfoot
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York, United Kingdom
| | - Eve Roman
- Epidemiology and Cancer Statistics Group, Department of Health Sciences, University of York, York, United Kingdom
| | - Sally E. Kinsey
- Department of Paediatric and Adolescent Haematology and Oncology, Leeds General Infirmary, Leeds, United Kingdom
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, United Kingdom
| | - Eamonn Sheridan
- Leeds Institute of Molecular Medicine, University of Leeds, Leeds, United Kingdom
| | - Pamela D. Thompson
- Paediatric and Familial Cancer, Institute of Cancer Sciences, Manchester, United Kingdom
| | - Per Hoffmann
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Human Genomics Research Group, Department of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Markus M. Nöthen
- Institute of Human Genetics, University of Bonn, Bonn, Germany
- Genomic Imaging Group, Institute of Neuroscience and Medicine (INM-1), Research Centre Juelich, Juelich, Germany
| | | | - Lewin Eisele
- Institute for Medical Informatics, Biometry and Epidemiology, University Hospital Essen, University of Duisburg–Essen, Essen, Germany
| | - Claus R. Bartram
- Institute of Human Genetics, University of Heidelberg, Heidelberg, Germany
| | - Martin Schrappe
- General Paediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Mel Greaves
- Haemato-Oncology Research Unit, Division of Molecular Pathology, Institute of Cancer Research, Sutton, Surrey, United Kingdom
| | - Kari Hemminki
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre, Heidelberg, Germany
- Center for Primary Health Care Research, Lund University, Malmö, Sweden
| | - Christine J. Harrison
- Northern Institute for Cancer Research, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Martin Stanulla
- General Paediatrics, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Richard S. Houlston
- Division of Genetics and Epidemiology, The Institute of Cancer Research, Sutton, Surrey, United Kingdom
| |
Collapse
|
14
|
Xu H, Zhang H, Yang W, Yadav R, Morrison AC, Qian M, Devidas M, Liu Y, Perez-Andreu V, Zhao X, Gastier-Foster JM, Lupo PJ, Neale G, Raetz E, Larsen E, Bowman WP, Carroll WL, Winick N, Williams R, Hansen T, Holm JC, Mardis E, Fulton R, Pui CH, Zhang J, Mullighan CG, Evans WE, Hunger SP, Gupta R, Schmiegelow K, Loh ML, Relling MV, Yang JJ. Inherited coding variants at the CDKN2A locus influence susceptibility to acute lymphoblastic leukaemia in children. Nat Commun 2015; 6:7553. [PMID: 26104880 PMCID: PMC4544058 DOI: 10.1038/ncomms8553] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 05/20/2015] [Indexed: 02/05/2023] Open
Abstract
There is increasing evidence from genome-wide association studies for a strong inherited genetic basis of susceptibility to acute lymphoblastic leukaemia (ALL) in children, yet the effects of protein-coding variants on ALL risk have not been systematically evaluated. Here we show a missense variant in CDKN2A associated with the development of ALL at genome-wide significance (rs3731249, P=9.4 × 10(-23), odds ratio=2.23). Functional studies indicate that this hypomorphic variant results in reduced tumour suppressor function of p16(INK4A), increases the susceptibility to leukaemic transformation of haematopoietic progenitor cells, and is preferentially retained in ALL tumour cells. Resequencing the CDKN2A-CDKN2B locus in 2,407 childhood ALL cases reveals 19 additional putative functional germline variants. These results provide direct functional evidence for the influence of inherited genetic variation on ALL risk, highlighting the important and complex roles of CDKN2A-CDKN2B tumour suppressors in leukaemogenesis.
Collapse
Affiliation(s)
- Heng Xu
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Department of Laboratory Medicine, National Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hui Zhang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Department of Pediatrics, The first affiliated hospital of Guangzhou Medical University, Guangzhou, Guangdong 510120, China
| | - Wenjian Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Rachita Yadav
- Centre for Biological Sequence Analysis, The Technical University of Denmark, Kgs, Lyngby DK-2800, Denmark
| | - Alanna C. Morrison
- Department of Epidemiology, Human Genetics, and Environmental Sciences, School of Public Health, University of Texas Health Science Center, Houston, Texas 77030, USA
| | - Maoxiang Qian
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Meenakshi Devidas
- Department of Biostatistics, Epidemiology and Health Policy Research, College of Medicine, University of Florida, Gainesville, Florida 32610, USA
| | - Yu Liu
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Virginia Perez-Andreu
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Xujie Zhao
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Julie M. Gastier-Foster
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, and Departments of Pathology and Pediatrics, Ohio State University College of Medicine, Columbus, Ohio 43205, USA
| | - Philip J. Lupo
- Department of Pediatrics, Texas Children's Cancer Center, Baylor College of Medicine, Houston, Texas 77030, USA
| | - Geoff Neale
- Hartwell Center for Bioinformatics & Biotechnology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Elizabeth Raetz
- Huntsman Cancer Institute, The University of Utah, Salt Lake City, Utah 84112, USA
| | - Eric Larsen
- Maine Children's Cancer Program, Scarborough, Maine 04074, USA
| | - W. Paul Bowman
- Cook Children's Medical Center, Ft. Worth, Texas 38754, USA
| | - William L. Carroll
- Pediatric Oncology, Cancer Institute New York University, New York City, New York 10016, USA
| | - Naomi Winick
- Pediatric Hematology/Oncology, University of Texas Southwestern Medical Center, Dallas, Texas 75235, USA
| | | | - Torben Hansen
- The Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen DK-2200, Denmark
| | - Jens-Christian Holm
- Department of Pediatrics, The Children's Obesity Clinic, Copenhagen University Hospital Holbaek, Holbaek DK-4300, Denmark
| | - Elaine Mardis
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri 63108, USA
| | - Robert Fulton
- McDonnell Genome Institute, Washington University School of Medicine, St Louis, Missouri 63108, USA
| | - Ching-Hon Pui
- Hematological Malignancies Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Jinghui Zhang
- Department of Computational Biology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Charles G. Mullighan
- Hematological Malignancies Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Department of Pathology, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - William E. Evans
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Hematological Malignancies Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Stephen P. Hunger
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania 19104, USA
| | - Ramneek Gupta
- Centre for Biological Sequence Analysis, The Technical University of Denmark, Kgs, Lyngby DK-2800, Denmark
| | - Kjeld Schmiegelow
- Department of Paediatrics and Adolescent Medicine, The Juliane Marie Centre, The University Hospital Rigshospitalet, and the Institute of Clinical Medicine, Faculty of Health, University of Copenhagen, Copenhagen DK-2100, Denmark
| | - Mignon L. Loh
- Department of Pediatrics, Benioff Children's Hospital and the Helen Diller Family Comprehensive Cancer Center, University of California at San Francisco, San Francisco, California 94115, USA
| | - Mary V. Relling
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Hematological Malignancies Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| | - Jun J. Yang
- Department of Pharmaceutical Sciences, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
- Hematological Malignancies Program, Comprehensive Cancer Center, St. Jude Children's Research Hospital, Memphis, Tennessee 38105, USA
| |
Collapse
|
15
|
Scaini MC, Minervini G, Elefanti L, Ghiorzo P, Pastorino L, Tognazzo S, Agata S, Quaggio M, Zullato D, Bianchi-Scarrà G, Montagna M, D'Andrea E, Menin C, Tosatto SCE. CDKN2A unclassified variants in familial malignant melanoma: combining functional and computational approaches for their assessment. Hum Mutat 2014; 35:828-40. [PMID: 24659262 DOI: 10.1002/humu.22550] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Accepted: 03/03/2014] [Indexed: 01/03/2023]
Abstract
CDKN2A codes for two oncosuppressors by alternative splicing of two first exons: p16INK4a and p14ARF. Germline mutations are found in about 40% of melanoma-prone families, and most of them are missense mutations mainly affecting p16INK4a. A growing number of p16INK4a variants of uncertain significance (VUS) are being identified but, unless their pathogenic role can be demonstrated, they cannot be used for identification of carriers at risk. Predicting the effect of these VUS by either a "standard" in silico approach, or functional tests alone, is rather difficult. Here, we report a protocol for the assessment of any p16INK4a VUS, which combines experimental and computational tools in an integrated approach. We analyzed p16INK4a VUS from melanoma patients as well as variants derived through permutation of conserved p16INK4a amino acids. Variants were expressed in a p16INK4a-null cell line (U2-OS) and tested for their ability to block proliferation. In parallel, these VUS underwent in silico prediction analysis and molecular dynamics simulations. Evaluation of in silico and functional data disclosed a high agreement for 15/16 missense mutations, suggesting that this approach could represent a pilot study for the definition of a protocol applicable to VUS in general, involved in other diseases, as well.
Collapse
Affiliation(s)
- Maria Chiara Scaini
- Immunology and Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Kurian AW, Hare EE, Mills MA, Kingham KE, McPherson L, Whittemore AS, McGuire V, Ladabaum U, Kobayashi Y, Lincoln SE, Cargill M, Ford JM. Clinical evaluation of a multiple-gene sequencing panel for hereditary cancer risk assessment. J Clin Oncol 2014; 32:2001-9. [PMID: 24733792 PMCID: PMC4067941 DOI: 10.1200/jco.2013.53.6607] [Citation(s) in RCA: 382] [Impact Index Per Article: 38.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
PURPOSE Multiple-gene sequencing is entering practice, but its clinical value is unknown. We evaluated the performance of a customized germline-DNA sequencing panel for cancer-risk assessment in a representative clinical sample. METHODS Patients referred for clinical BRCA1/2 testing from 2002 to 2012 were invited to donate a research blood sample. Samples were frozen at -80° C, and DNA was extracted from them after 1 to 10 years. The entire coding region, exon-intron boundaries, and all known pathogenic variants in other regions were sequenced for 42 genes that had cancer risk associations. Potentially actionable results were disclosed to participants. RESULTS In total, 198 women participated in the study: 174 had breast cancer and 57 carried germline BRCA1/2 mutations. BRCA1/2 analysis was fully concordant with prior testing. Sixteen pathogenic variants were identified in ATM, BLM, CDH1, CDKN2A, MUTYH, MLH1, NBN, PRSS1, and SLX4 among 141 women without BRCA1/2 mutations. Fourteen participants carried 15 pathogenic variants, warranting a possible change in care; they were invited for targeted screening recommendations, enabling early detection and removal of a tubular adenoma by colonoscopy. Participants carried an average of 2.1 variants of uncertain significance among 42 genes. CONCLUSION Among women testing negative for BRCA1/2 mutations, multiple-gene sequencing identified 16 potentially pathogenic mutations in other genes (11.4%; 95% CI, 7.0% to 17.7%), of which 15 (10.6%; 95% CI, 6.5% to 16.9%) prompted consideration of a change in care, enabling early detection of a precancerous colon polyp. Additional studies are required to quantify the penetrance of identified mutations and determine clinical utility. However, these results suggest that multiple-gene sequencing may benefit appropriately selected patients.
Collapse
Affiliation(s)
- Allison W Kurian
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Emily E Hare
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Meredith A Mills
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Kerry E Kingham
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Lisa McPherson
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Alice S Whittemore
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Valerie McGuire
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Uri Ladabaum
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Yuya Kobayashi
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Stephen E Lincoln
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - Michele Cargill
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA
| | - James M Ford
- Allison W. Kurian, Meredith A. Mills, Kerry E. Kingham, Lisa McPherson, Alice S. Whittemore, Valerie McGuire, Uri Ladabaum, James M. Ford, Stanford University School of Medicine, Stanford; Emily E. Hare, Yuya Kobayashi, Stephen E. Lincoln, Michele Cargill, InVitae, San Francisco, CA.
| |
Collapse
|
17
|
Fan M, Pfeffer SR, Lynch HT, Cassidy P, Leachman S, Pfeffer LM, Kopelovich L. Altered transcriptome signature of phenotypically normal skin fibroblasts heterozygous for CDKN2A in familial melanoma: relevance to early intervention. Oncotarget 2013; 4:128-41. [PMID: 23371019 PMCID: PMC3702213 DOI: 10.18632/oncotarget.786] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Familial melanoma (FM) is a dominantly heritable cancer that is associated with mutations in the tumor suppressor CDKN2A/p16. In FM, a single inherited “hit” occurs in every somatic cell, enabling interrogation of cultured normal skin fibroblasts (SFs) from FM gene carriers as surrogates for the cell of tumor origin, namely the melanocyte. We compared the gene expression profile of SFs from FM individuals with two distinct CDKN2A/p16 mutations (V126D-p16 and R87P-p16) with the gene expression profile of SFs from age-matched individuals without p16 mutations and with no family history of melanoma. We show an altered transcriptome signature in normal SFs bearing a single-hit inherited mutation in the CDKN2A/p16 gene, wherein some of these abnormal alterations recapitulate changes observed in the corresponding cancer. Significantly, the extent of the alterations is mutation-site specific with the R87P-p16 mutation being more disruptive than the V126D-p16 mutation. We also examined changes in gene expression after exposure to ultraviolet (UV) radiation to define potential early biomarkers triggered by sun exposure. UV treatment of SFs from FM families induces distinct alterations in genes related to cell cycle regulation and DNA damage responses that are also reported to be dysregulated in melanoma. Importantly, these changes were diametrically opposed to UV-induced changes in SF from normal controls. We posit that changes identified in the transcriptome of SF from FM mutation carriers represent early events critical for melanoma development. As such, they may serve as specific biomarkers of increased risk as well as molecular targets for personalized prevention strategies in high-risk populations.
Collapse
Affiliation(s)
- Meiyun Fan
- Department of Pathology and Laboratory Medicine, and the Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Bakos RM, Besch R, Zoratto GG, Godinho JM, Mazzotti NG, Ruzicka T, Bakos L, Santos SE, Ashton-Prolla P, Berking C, Giugliani R. The CDKN2A p.A148T variant is associated with cutaneous melanoma in Southern Brazil. Exp Dermatol 2011; 20:890-3. [PMID: 21895773 DOI: 10.1111/j.1600-0625.2011.01332.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Several germline mutations and sequence variants in cancer predisposition genes have been described. Among these, the CDKN2A p.A148T variant appears to be frequent in patients with melanoma, at least in certain ethnic groups. In this case-control study, we evaluated 127 patients with cutaneous melanoma and 128 controls from Southern Brazil, the region with the highest melanoma incidence rates in the country. Using PCR-RFLP, we demonstrate that CDKN2A p.A148T variant was significantly more frequent in patients with melanoma than in controls (12.6% vs 3.9%; P=0.009). There was no association between presence of the polymorphism and tumor thickness, site of the primary tumor, melanoma subtype, age at diagnosis, quantitative and qualitative number of nevi. Patients with a positive family of history for other cancers were particularly prone to carry the CDKN2A p.A148T allele. All patients with p.A148T-positive melanoma reported European ancestry, especially German, and this was confirmed using a panel of ancestry-informative INDELs. Our data suggest that CDKN2A p.A148T is a melanoma susceptibility allele in Southern Brazil and is particularly common in patients with melanoma of predominantly European ancestry.
Collapse
Affiliation(s)
- Renato M Bakos
- Dermatology Service, Hospital de Clínicas de Porto Alegre (HCPA), Brazil
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Choi JW, Lee JH, Kim YS. Frequent upregulation of cyclin D1 and p16 expression with low Ki-67 scores in multinucleated giant cells. Pathobiology 2011; 78:233-7. [PMID: 21778791 DOI: 10.1159/000327359] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 03/15/2011] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND/AIMS Multinucleated giant cells are formed from the fusion of macrophages and are classified into foreign body-type giant cells (FBGCs), osteoclast-type giant cells (OCGCs) and Langhans-type giant cells (LHGCs). OCGCs display upregulated cyclin D1 expression with low Ki-67 activity. However, little is known about the expression of cell cycle regulators in the other types of multinucleated giant cells. We aimed to investigate the cell cycle status of multinucleated giant cells. METHODS The immunohistochemical expressions of cyclin D1, p16(INK4a) and Ki-67 were analyzed in a total of 127 cases showing multinucleated giant cells. RESULTS Cyclin D1 was overexpressed in 45 (88%) of 51 FBGC cases, 25 (86%) of 29 OCGC cases and 22 (47%) of 47 LHGC cases. p16(INK4a) showed diffuse nuclear and/or cytoplasmic overexpression in 45 (88%) of 51 FBGC cases, 27 (93%) of 29 OCGC cases and 24 (51%) of 47 LHGC cases. Ki-67 immunostaining was negative in almost all FBGC, OCGC and LHGC cases. CONCLUSION This study demonstrates that FBGCs and OCGCs frequently show upregulation of cyclin D1 and p16(INK4a) expression with low Ki-67 scores. This suggests that multinucleated giant cells are arrested in the G1/S cell cycle transition.
Collapse
Affiliation(s)
- Jung-Woo Choi
- Department of Pathology, Korea University Ansan Hospital, Ansan, Republic of Korea
| | | | | |
Collapse
|
20
|
McKenzie HA, Fung C, Becker TM, Irvine M, Mann GJ, Kefford RF, Rizos H. Predicting functional significance of cancer-associated p16(INK4a) mutations in CDKN2A. Hum Mutat 2010; 31:692-701. [PMID: 20340136 DOI: 10.1002/humu.21245] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Inherited mutations affecting the INK4a/ARF locus (CDKN2A) are associated with melanoma susceptibility in 40% of multiple case melanoma families. Over 60 different germline INK4a/ARF mutations have been detected in more than 190 families worldwide. The majority of these alterations are missense mutations affecting p16(INK4a), and only 25% of these have been functionally assessed. There is therefore a need for an accurate and rapid assay to determine the functional significance of p16(INK4a) mutations. We reviewed the performance of several in vivo functional assays that measure critical aspects of p16(INK4a) function, including subcellular location, CDK binding and cell cycle inhibition. In this report the function of 28 p16(INK4a) variants, many associated with melanoma susceptibility were compared. We show that assessment of CDK4 binding and subcellular localization can accurately and rapidly determine the functional significance of melanoma-associated p16(INK4a) mutations. p16(INK4a)-CDK6 binding affinity was unhelpful, as no disease-associated mutation showed reduced CDK6 affinity while maintaining the ability to bind CDK4. Likewise, in silico analyses did not contribute substantially, with only 12 of 25 melanoma-associated missense variants consistently predicted as deleterious. The ability to determine variant functional activity accurately would identify disease-associated mutations and facilitate effective genetic counselling of individuals at high risk of melanoma.
Collapse
Affiliation(s)
- Heather A McKenzie
- Westmead Institute for Cancer Research and Melanoma Institute of Australia, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead NSW 2145, Australia
| | | | | | | | | | | | | |
Collapse
|
21
|
Bisio A, Nasti S, Jordan JJ, Gargiulo S, Pastorino L, Provenzani A, Quattrone A, Queirolo P, Bianchi-Scarrà G, Ghiorzo P, Inga A. Functional analysis of CDKN2A/p16INK4a 5'-UTR variants predisposing to melanoma. Hum Mol Genet 2010; 19:1479-91. [PMID: 20093296 DOI: 10.1093/hmg/ddq022] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Germline CDKN2A mutations are observed in 20-50% of melanoma-prone families. We identified melanoma patients that were heterozygous for non-coding germline variants in the 5'-UTR of CDKN2A (c.-21C > T; c.-25C > T&c.-180G > A; c.-56G > T; c.-67G > C) and examined their impact on the p16(INK4a) 5'-UTR activity using two luciferase-based reporter vectors that differ in basal transcription level and that were transfected into the melanoma-derived WM266-4 and in the breast cancer-derived MCF7 cells. The wild-type 5'-UTR sequence, containing a reported SNP (c.-33G > C) and a known melanoma-predisposing mutation (c.-34G > T), was included as controls. Results revealed that the variants at -21 and -34 severely reduced the reporter activity. The variants at -56 and at -25&-180 exhibited a milder impact, while results with c.-67G > C were dependent on the plasmid type. Quantification of the luciferase mRNA indicated that the effects of the variants were mainly post-transcriptional. Using a bicistronic dual-luciferase reporter plasmid, we confirmed that c.-21C > T and c.-34G > T had a severe negative impact in both cell lines. We also applied a polysomal profiling technique to samples heterozygous for the 5'-UTR variants, including patient-derived lymphoblasts. Analysis of allelic imbalance indicated that in addition to the c.-21C > T variant, the c.-56T > G and c.-67G > C variants also reduced mRNA translation efficiency. Overall, our results suggest that the c.-21C > T sequence variant is a melanoma-predisposing mutation. The c.-25C > T&c.-180G > A and particularly the c.-56G > T variants showed a range of intermediate functional defects in the different assays, and were not observed in the control population. We propose that these variants should be considered as potential mutations.
Collapse
Affiliation(s)
- Alessandra Bisio
- Unit of Molecular Mutagenesis and DNA Repair, National Institute for Cancer Research IST, 16132 Genoa, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Scaini MC, Rossi E, de Siqueira Torres PLA, Zullato D, Callegaro M, Casella C, Quaggio M, Agata S, Malacrida S, Chiarion-Sileni V, Vecchiato A, Alaibac M, Montagna M, Mann GJ, Menin C, D'Andrea E. Functional impairment of p16(INK4A) due to CDKN2A p.Gly23Asp missense mutation. Mutat Res 2009; 671:26-32. [PMID: 19712690 DOI: 10.1016/j.mrfmmm.2009.08.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 08/11/2009] [Accepted: 08/12/2009] [Indexed: 05/28/2023]
Abstract
The CDKN2A locus encodes for two distinct tumor suppressor proteins, p16(INK4A) and p14(ARF), involved in cell cycle regulation. CDKN2A germline mutations have been associated with familial predisposition to melanoma and other tumor types. Besides bona-fide pathogenic mutations, many sequence variants have been identified, but their effect is not well known. We detected the p.Gly23Asp missense mutation in one of the two tested melanoma patients of a family with three melanoma cases. Even though the mutated amino acid is located in a conserved domain that specifically binds to and blocks the function of CDK4/6, its lack of segregation with disease suggested a series of functional assays to discriminate between a pathogenic variant and a neutral polymorphism. The effect of this mutation has been investigated exploiting four p16(INK4A) properties: its ability (i) to bind CDK4, (ii) to inhibit pRb phosphorylation, (iii) to evenly localize in the cell, and (iv) to cause cell cycle arrest. The mutant protein properties were evaluated transfecting three different cell lines (U2-OS and NM-39, both p16-null, and SaOS 2, p53 and pRb-null) with plasmids expressing either p16(wt), p16(23Asp), or the p16(32Pro) pathogenic variant. We found that p16(23Asp) was less efficient than p16(wt) in CDK4 binding, in inhibiting pRb phosphorylation, in inducing G1 cell cycle arrest; moreover, its pattern of distribution throughout the cell was suggestive of protein aggregation, thus assessing a pathogenic role for p16(23Asp) in familial melanoma.
Collapse
Affiliation(s)
- Maria Chiara Scaini
- Section of Oncology, Department of Oncology and Surgical Sciences, University of Padova, via Gattamelata, 64, I-35128 Padova, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
McKenzie H, Becker TM, Scurr LL, Kefford RF, Rizos H. Wild type and melanoma-associated mutant p16(IN4a) proteins do not oligomerize in vivo. Pigment Cell Melanoma Res 2009; 22:131-3. [PMID: 19154236 DOI: 10.1111/j.1755-148x.2008.00530.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
24
|
Couch FJ, Rasmussen LJ, Hofstra R, Monteiro ANA, Greenblatt MS, de Wind N. Assessment of functional effects of unclassified genetic variants. Hum Mutat 2008; 29:1314-26. [PMID: 18951449 PMCID: PMC2771414 DOI: 10.1002/humu.20899] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Inherited predisposition to disease is often linked to reduced activity of a disease associated gene product. Thus, quantitation of the influence of inherited variants on gene function can potentially be used to predict the disease relevance of these variants. While many disease genes have been extensively characterized at the functional level, few assays based on functional properties of the encoded proteins have been established for the purpose of predicting the contribution of rare inherited variants to disease. Much of the difficulty in establishing predictive functional assays stems from the technical complexity of the assays. However, perhaps the most challenging aspect of functional assay development for clinical testing purposes is the absolute requirement for validation of the sensitivity and specificity of the assays and the determination of positive predictive values (PPVs) and negative predictive values (NPVs) of the assays relative to a "gold standard" measure of disease predisposition. In this commentary, we provide examples of some of the functional assays under development for several cancer predisposition genes (BRCA1, BRCA2, CDKN2A, and mismatch repair [MMR] genes MLH1, MSH2, MSH6, and PMS2) and present a detailed review of the issues associated with functional assay development. We conclude that validation is paramount for all assays that will be used for clinical interpretation of inherited variants of any gene, but note that in certain circumstances information derived from incompletely validated assays may be valuable for classification of variants for clinical purposes when used to supplement data derived from other sources.
Collapse
Affiliation(s)
- Fergus J Couch
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota 55905, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Mihic-Probst D, Kuster A, Kilgus S, Bode-Lesniewska B, Ingold-Heppner B, Leung C, Storz M, Seifert B, Marino S, Schraml P, Dummer R, Moch H. Consistent expression of the stem cell renewal factor BMI-1 in primary and metastatic melanoma. Int J Cancer 2007; 121:1764-70. [PMID: 17597110 DOI: 10.1002/ijc.22891] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Stem cell-like cells have recently been identified in melanoma cell lines, but their relevance for melanoma pathogenesis is controversial. To characterize the stem cell signature of melanoma, expression of stem cell markers BMI-1 and nestin was studied in 64 cutaneous melanomas, 165 melanoma metastases as well as 53 melanoma cell lines. Stem cell renewal factor BMI-1 is a transcriptional repressor of the Ink4a/Arf locus encoding p16(ink4a) and p14(Arf). Increased nuclear BMI-1 expression was detectable in 41 of 64 (64%) primary melanomas, 117 of 165 melanoma metastases (71%) and 15 of 53 (28%) melanoma cell lines. High nestin expression was observed in 14 of 56 primary melanomas (25%), 84 of 165 melanoma metastases (50%) and 21 of 53 melanoma cell lines (40%). There was a significant correlation between BMI-1 and nestin expression in cell lines (p = 0.001) and metastases (p = 0.02). These data indicate that cells in primary melanomas and their metastases may have stem cell properties. Cell lines obtained from melanoma metastases showed a significant higher BMI-1 expression compared to cell lines from primary melanoma (p = 0.001). Further, primary melanoma lacking lymphatic metastases at presentation (pN0, n = 40) was less frequently BMI-1 positive than melanomas presenting with lymphatic metastases (pN1; n = 24; 52% versus 83%; p = 0.01). Therefore, BMI-1 expression appears to induce a metastatic tendency. Because BMI-1 functions as a transcriptional repressor of the Ink4a/Arf locus, p16(ink4a) and p14(Arf) expression was also analyzed. A high BMI-1/low p16(ink4a) expression pattern was a significant predictor of metastasis by means of logistic regression analysis (p = 0.005). This suggests that BMI-1 mediated repression of p16(ink4a) may contribute to an increased aggressive behavior of stem cell-like melanoma cells.
Collapse
Affiliation(s)
- Daniela Mihic-Probst
- Department of Pathology, Institute of Surgical Pathology, University Hospital Zurich, Zurich, Switzerland.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chan PA, Duraisamy S, Miller PJ, Newell JA, McBride C, Bond JP, Raevaara T, Ollila S, Nyström M, Grimm AJ, Christodoulou J, Oetting WS, Greenblatt MS. Interpreting missense variants: comparing computational methods in human disease genes CDKN2A, MLH1, MSH2, MECP2, and tyrosinase (TYR). Hum Mutat 2007; 28:683-93. [PMID: 17370310 DOI: 10.1002/humu.20492] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The human genome contains frequent single-basepair variants that may or may not cause genetic disease. To characterize benign vs. pathogenic missense variants, numerous computational algorithms have been developed based on comparative sequence and/or protein structure analysis. We compared computational methods that use evolutionary conservation alone, amino acid (AA) change alone, and a combination of conservation and AA change in predicting the consequences of 254 missense variants in the CDKN2A (n = 92), MLH1 (n = 28), MSH2 (n = 14), MECP2 (n = 30), and tyrosinase (TYR) (n = 90) genes. Variants were validated as either neutral or deleterious by curated locus-specific mutation databases and published functional data. All methods that use evolutionary sequence analysis have comparable overall prediction accuracy (72.9-82.0%). Mutations at codons where the AA is absolutely conserved over a sufficient evolutionary distance (about one-third of variants) had a 91.6 to 96.8% likelihood of being deleterious. Three algorithms (SIFT, PolyPhen, and A-GVGD) that differentiate one variant from another at a given codon did not significantly improve predictive value over conservation score alone using the BLOSUM62 matrix. However, when all four methods were in agreement (62.7% of variants), predictive value improved to 88.1%. These results confirm a high predictive value for methods that use evolutionary sequence conservation, with or without considering protein structural change, to predict the clinical consequences of missense variants. The methods can be generalized across genes that cause different types of genetic disease. The results support the clinical use of computational methods as one tool to help interpret missense variants in genes associated with human genetic disease.
Collapse
Affiliation(s)
- Philip A Chan
- Vermont Cancer Center, University of Vermont, Burlington, Vermont, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Orlow I, Begg CB, Cotignola J, Roy P, Hummer AJ, Clas BA, Mujumdar U, Canchola R, Armstrong BK, Kricker A, Marrett LD, Millikan RC, Gruber SB, Anton-Culver H, Zanetti R, Gallagher RP, Dwyer T, Rebbeck TR, Kanetsky PA, Wilcox H, Busam K, From L, Berwick M. CDKN2A germline mutations in individuals with cutaneous malignant melanoma. J Invest Dermatol 2007; 127:1234-43. [PMID: 17218939 DOI: 10.1038/sj.jid.5700689] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Cyclin-dependent kinase inhibitor type 2A (CDKN2A) has been identified as a major melanoma susceptibility gene based on the presence of germline mutations in high-risk melanoma families. In this study, we sought to identify and characterize the spectrum of CDKN2A mutations affecting p16 inhibitor of cyclin-dependent kinase type 4 (INK4a) in individuals with melanoma using a population-based study design. DNA samples from 1189 individuals with incident multiple primary melanoma (MPM) and 2424 with incident single primary melanoma unselected for family history of melanoma were available for screening of CDKN2A (p16INK4a) mutations. Variants were classified for functional impact based on intragenic position, existing functional data, sequence, and structural analysis. The impact of individual mutations and functional groupings was assessed by comparing frequencies in cases of MPM versus cases with a single first primary melanoma, and by comparing the reported incidence rates in first-degree relatives. Our results show that mutations occur infrequently in these high-risk groups, and that they occur mainly in exons 1alpha and 2. Rare coding variants with putative functional impact are observed to increase substantially the risk of melanoma. With the exception of the variant in position -34 of CDKN2A of known functional consequence, the remaining rare variants in the non-coding region have no apparent impact on risk.
Collapse
Affiliation(s)
- Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, New York 10021, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Mihic-Probst D, Mnich CD, Oberholzer PA, Seifert B, Sasse B, Moch H, Dummer R. p16 expression in primary malignant melanoma is associated with prognosis and lymph node status. Int J Cancer 2006; 118:2262-8. [PMID: 16331607 DOI: 10.1002/ijc.21608] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Lymph node (LN) status is an important prognostic factor in melanoma patients. p16 expression and proliferation rate (MIB-1) of primary melanomas have been suggested as a marker of metastatic potential. In this study, the correlation of p16 expression and the proliferation rate (MIB-1) with LN status and tumor-specific survival was investigated in primary melanomas. MIB-1 and p16 expression were analyzed by immunohistochemistry in 64 patients with primary cutaneous melanoma. Thirty four nevi were used as control. All patients underwent sentinel lymph node staging. Three different p16 staining patterns were observed: a combination of nuclear and cytoplasmic staining, only cytoplasmic staining and absence of p16 expression. All 34 nevi displayed a nuclear and cytoplasmic p16 staining, whereas p16 was negative in 14 of 64 (22%) melanomas. The level of p16 expression gradually decreased from benign nevi to melanoma without metastasis to melanoma with metastasis. There was a significant correlation between cytoplasmic p16 expression and absence of metastasis (p < 0.05). Death of disease correlated with absence of p16 immunostaining (p = 0.01). MIB-1 expression was not associated with survival. These results confirm the relevance of p16 expression as a prognostic marker in melanoma patients. In addition, it was shown that cytoplasmic immunostaining for p16 in primary melanoma might serve as a predictor of the LN status. Therefore, immunohistochemical evaluation for p16 expression is of potential value for treatment planning in melanoma surgery.
Collapse
Affiliation(s)
- Daniela Mihic-Probst
- Department of Pathology, Institute of Surgical Pathology, University Hospital, Zürich, Switzerland.
| | | | | | | | | | | | | |
Collapse
|
29
|
Chen J, Huang X, Halicka D, Brodsky S, Avram A, Eskander J, Bloomgarden NA, Darzynkiewicz Z, Goligorsky MS. Contribution of p16INK4aand p21CIP1pathways to induction of premature senescence of human endothelial cells: permissive role of p53. Am J Physiol Heart Circ Physiol 2006; 290:H1575-86. [PMID: 16243918 DOI: 10.1152/ajpheart.00364.2005] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
We have previously found that nonenzymatically glycated collagen I (GC), mimicking diabetic microenvironment, can induce senescent phenotype in early passage human umbilical vein endothelial cells (HUVECs). In the present study, we explored the functional involvement of cell cycle checkpoint pathways in initiating GC-induced premature endothelial cell senescence. When compared with native collagen, early passage HUVECs showed increased p53, p21CIP1(p21), and p16INK4a(p16) mRNA expression after exposure to GC. Twenty-four hours after transfection of p16, p21, and p53-enhanced green fluorescent protein (EGFP) recombinant plasmids, HUVECs entered G1-phase cell cycle arrest. By days 3 and 5, HUVECs transfected with p16-EGFP showed an increased proportion of senescent cells, and this increase was more prominent in the GFP-positive cell population, which exhibited 68% of senescent cells. Transfection of p21 also induced senescence but only by day 5. Cotransfection of p16 and p21 showed no additive effect. Transfection of p21 or p53 induced apoptosis in HUVECs. Next, we suppressed endogenous p53, p21, p16, or retinoblastoma (Rb) gene expression through small interference RNA strategy and investigated their influence in p16- and p21-initiated endothelial cell senescence. Analysis indicated that suppression of p53 expression can abolish senescence induced by p16 overexpression. Paradoxically, this effect was not observed when p21 was suppressed. On the other hand, suppression of Rb eliminated senescence initiated by either p16 or p21 overexpression. In summary, the p53/p21 pathway is mainly responsible for GC-induced apoptosis, but the coordinated activation of the p53/p21 and p16 pathway is responsible for GC-induced endothelial cell senescence through a Rb-dependent mechanism.
Collapse
Affiliation(s)
- Jun Chen
- Department of Medicine, New York Medical College, Valhalla, NY 10595, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Nilsson K, Landberg G. Subcellular localization, modification and protein complex formation of the cdk-inhibitor p16 in Rb-functional and Rb-inactivated tumor cells. Int J Cancer 2005; 118:1120-5. [PMID: 16161044 DOI: 10.1002/ijc.21466] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The cdk-inhibitor p16 is a tumor suppressor gene that is inactivated in many forms of cancer. Despite numerous studies, the exact mechanism of regulation of p16 has not been clarified, although the status of retinoblastoma (Rb) seems to be one important factor that influences the p16 expression. The specificity and validity of cytoplasmic localization of p16 observed in some tumors has further been questioned. Here, by subcellular fractionation of Rb-functional and Rb-inactivated cell lines, we show that p16 indeed is expressed in the cytoplasm as well as in the nucleus. Post translational modifications of p16 in different subcellular compartments as well as its capacity to form protein complexes were further delineated. Two dimensional gel electrophoresis showed that two forms of p16 appeared in the cytoplasm, while only one form was detected in the nucleus. Samples of basal cell carcinoma and squamous cell carcinoma of the skin with either functional or non-functional Rb also exhibited at least two forms of p16. In addition, cytoplasmic p16 bound cyclin dependent kinase (cdk)4/6, potentially indicating that p16 could have a function in the cytoplasm.
Collapse
Affiliation(s)
- Kristina Nilsson
- Division of Pathology, Department of Laboratory Medicine, Lund University, Malmö University Hospital, S-205 02 Malmö, Sweden
| | | |
Collapse
|
31
|
Becker TM, Rizos H, de la Pena A, Leclercq IA, Woodruff S, Kefford RF, Mann GJ. Impaired inhibition of NF-kappaB activity by melanoma-associated p16INK4a mutations. Biochem Biophys Res Commun 2005; 332:873-9. [PMID: 15913553 DOI: 10.1016/j.bbrc.2005.05.032] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2005] [Accepted: 05/06/2005] [Indexed: 11/19/2022]
Abstract
The cyclin-dependent kinase inhibitor p16INK4a has been identified as tumor suppressor and melanoma predisposition gene. While its cell cycle inhibitory ability is important in protecting cells from uncontrolled growth and possible tumor formation, other functions of p16INK4a are likely to contribute to its nature as a tumor suppressor. p16INK4a binding and inhibition of the transcription factor NF-kappaB has been shown and is consistent with the reports of abnormally increased NF-kappaB activity in various cancers including melanoma. Here, we present evidence that wild type p16INK4a binds to the NF-kappaB subunit RelA more efficiently than melanoma-associated p16INK4a mutations. Furthermore, whereas wild type p16INK4a strongly inhibits NF-kappaB transcriptional activity, a subset of melanoma-associated p16INK4a mutants show reduced NF-kappaB inhibitory function. p16INK4a does not affect NF-kappaB nuclear translocation or DNA binding, suggesting a mechanism that reduces NF-kappaB transactivation activity.
Collapse
Affiliation(s)
- T M Becker
- Westmead Institute for Cancer Research, University of Sydney, Westmead Millennium Institute, Westmead, NSW 2145, Australia.
| | | | | | | | | | | | | |
Collapse
|
32
|
Mosavi LK, Cammett TJ, Desrosiers DC, Peng ZY. The ankyrin repeat as molecular architecture for protein recognition. Protein Sci 2005; 13:1435-48. [PMID: 15152081 PMCID: PMC2279977 DOI: 10.1110/ps.03554604] [Citation(s) in RCA: 638] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The ankyrin repeat is one of the most frequently observed amino acid motifs in protein databases. This protein-protein interaction module is involved in a diverse set of cellular functions, and consequently, defects in ankyrin repeat proteins have been found in a number of human diseases. Recent biophysical, crystallographic, and NMR studies have been used to measure the stability and define the various topological features of this motif in an effort to understand the structural basis of ankyrin repeat-mediated protein-protein interactions. Characterization of the folding and assembly pathways suggests that ankyrin repeat domains generally undergo a two-state folding transition despite their modular structure. Also, the large number of available sequences has allowed the ankyrin repeat to be used as a template for consensus-based protein design. Such projects have been successful in revealing positions responsible for structure and function in the ankyrin repeat as well as creating a potential universal scaffold for molecular recognition.
Collapse
Affiliation(s)
- Leila K Mosavi
- MC3305, Department of Molecular, Microbial, and Structural Biology, University of Connecticut Health Center, 263 Farmington Avenue, Farmington, CT 06032, USA
| | | | | | | |
Collapse
|
33
|
Becker TM, Ayub AL, Kefford RF, Mann GJ, Rizos H. The melanoma-associated 24 base pair duplication in p16INK4a is functionally impaired. Int J Cancer 2005; 117:569-73. [PMID: 15945100 DOI: 10.1002/ijc.21265] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Melanoma-associated germline mutations affecting the tumor suppressor and cyclin-dependent kinase (CDK) inhibitor, CDKN2A/p16INK4a, have been identified in over 100 melanoma-prone families worldwide. To predict the melanoma risk for carriers of specific mutations, mutant p16INK4a can be tested in biochemical and cellular assays. In most cases, p16INK4a mutations with predicted disease relation, due to segregation with melanoma, are functionally impaired in such assays. The N-terminal 24 base pair duplication of CDKN2A, however, encodes a p16INK4a variant previously shown to have wild-type function, despite segregating with melanoma in at least 5 melanoma families. To clarify whether the duplication mutation has a cell cycle regulatory defect or behaves like wild-type p16INK4a, we reanalyzed the cell cycle-inhibitory activity of this mutation. Stable cell clones of the p16-null WMM1175 melanoma cell line inducible for ectopic p16INK4a were used in this study. In these cells, p16INK4a expression can be controlled at physiologic levels. Our results show that in comparison to wild-type p16INK4a, the duplication mutant induced weaker S-phase inhibition and cells expressing this mutant form of p16INK4a retained colony formation ability. We also show that the cell cycle-regulatory defect of the p16INK4a duplication mutant was associated with decreased inhibition of pRb phosphorylation even though it retained significant binding to CDK4.
Collapse
Affiliation(s)
- Therese M Becker
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, New South Wales, Australia.
| | | | | | | | | |
Collapse
|
34
|
Ghiorzo P, Mantelli M, Gargiulo S, Gramigni C, Pastorino L, Banelli B, Villaggio B, Coccia MC, Sementa AR, Garrè C, Bianchi-Scarrà G. Inverse correlation between p16INK4A expression and NF-kappaB activation in melanoma progression. Hum Pathol 2004; 35:1029-37. [PMID: 15297971 DOI: 10.1016/j.humpath.2004.02.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Expression of p16INK4A, the product of the melanoma susceptibility gene CDKN2A, has been shown to decrease in correlation with tumor progression. P16INK4A is a key regulator of cell-cycle function, and likely interacts with a variety of targets alongside cyclin-dependent kinases (CDKs). One such target is nuclear factor KB (NF-kappaB), a pleiotropic transcription factor that plays a crucial role in apoptosis, oncogenesis and cell cycle control. NF-kappaB p65 has been shown to be activated in melanoma cell lines but few studies decribe its expression in the tissue. In the present study we focused on synchronous expression of p16INK4A and NF-kappaB p65 and their functional activation in melanoma cell lines and biopsy tissue. Activation of NF-kappaB p65, as observed by electrophoretic mobility shift assay in cell lines, was correlated with expression and cellular localization of the active and inactive forms of its inhibitor, IkappaB-alpha. In melanocytic lesions, p16INK4A and NF-kappaB p65 expression were inversely correlated with levels of the nuclear component of NF-kappaB p65 increasing from nevi to primary melanomas and metastases.
Collapse
Affiliation(s)
- Paola Ghiorzo
- Department of Oncology, Biology and Genetics, University of Genova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Scarrà GB, Testori A, Mihm M. What is new in melanoma research: genetics and epidemiology of melanoma in 2003? Review of a workshop held in Milan in May 2003. Melanoma Res 2004; 14:77-84. [PMID: 15057035 DOI: 10.1097/00008390-200404000-00001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
The various papers that were presented during this symposium touched on two of the most important aspects concerning melanoma because they dealt with epidemiology and genetics. Secondary prevention represents the most important step to cure melanoma patients once the disease has presented on a patient. It is clear that educational campaigns lead to greater awareness of the public and hence better prevention and earlier diagnosis. A second item is becoming more and more important in the field of prevention and early diagnosis, which is to understand the genetic basis of melanoma etiology and development. Studies concerning the familial clustering, for example, can lead to more careful monitoring of patients for other tumours and also lead to a basis for further research.
Collapse
|
36
|
Ghiorzo P, Villaggio B, Sementa AR, Hansson J, Platz A, Nicoló G, Spina B, Canepa M, Palmer JM, Hayward NK, Bianchi-Scarrà G. Expression and localization of mutant p16 proteins in melanocytic lesions from familial melanoma patients. Hum Pathol 2004; 35:25-33. [PMID: 14745721 DOI: 10.1016/j.humpath.2003.08.017] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Little is known about the correlation between the loss of p16 expression and tumor progression in familial melanoma; no systematic study has been conducted on p16 expression in melanocytic tumors from patients carrying germline CDKN2A mutations. We analyzed 98 early primary lesions from familial patients, previously tested for germline CDKN2A status, by quantitative immunohistochemistry using 3 p16 antibodies. We found that p16 expression was inversely correlated with tumor progression and was significantly lower in melanomas, including in situ lesions, than in nevi. Of other features analyzed, tumor thickness showed the most significant correlation with p16 levels. Lesions from mutation-negative patients displayed combined nuclear and cytoplasmic staining. However, some mutation-positive lesions (ie, G101W, 113insR, M53I, R24P, and 33ins24), including benign nevi, showed nuclear mislocalization, confirming previous studies suggesting that subcellular distribution indicates functional impairment of p16.
Collapse
Affiliation(s)
- Paola Ghiorzo
- Department of Oncology, Biology, and Genetics, University of Genova, Genova, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Kreimer-Erlacher H, Seidl H, Bäck B, Cerroni L, Kerl H, Wolf P. High frequency of ultraviolet mutations at the INK4a-ARF locus in squamous cell carcinomas from psoralen-plus-ultraviolet-A-treated psoriasis patients. J Invest Dermatol 2003; 120:676-82. [PMID: 12648234 DOI: 10.1046/j.1523-1747.2003.12085.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Squamous cell carcinomas in psoralen-plus-ultraviolet A (PUVA) treated patients frequently exhibit p53 tumor suppressor genes and Ha-ras protooncogenes that are mutated at dipyrimidine sites and carry the ultraviolet fingerprint (i.e., C-to-T or CC-to-TT transitions). To further broaden the knowledge of genetic mutations in PUVA-associated skin cancer, we used DNA sequencing analysis to study the mutational spectrum of the INK4a-ARF locus in 26 squamous cell carcinomas from 11 long-term PUVA-treated psoriasis patients and classified the mutations by origin (ultraviolet, ultraviolet and/or PUVA, or other). Nineteen INK4a-ARF missense/nonsense mutations were found in exons 1alpha, 1beta, and 2 in 11 of 26 squamous cell carcinomas (42%) from seven of 11 patients (64%). Eleven mutations (58%) were of the ultraviolet type; three (16%) were of the ultraviolet and/or PUVA type (i.e., C-to-T transitions at dipyrimidine sites opposite a 5'TpG sequence, a potential psoralen binding site); and five (26%) were of other type. Interestingly, 10 of 11 patients (91%) showed intron polymorphism C500G at the 3' untranslated region of exon 3. These data indicate that (i) INK4a-ARF mutations frequently occur in PUVA-associated squamous cell carcinomas; (ii) ultraviolet B radiation is the major cause of these mutations; and (iii) PUVA itself may play no direct role in development of most INK4a-ARF mutations.
Collapse
|
38
|
Greenblatt MS, Beaudet JG, Gump JR, Godin KS, Trombley L, Koh J, Bond JP. Detailed computational study of p53 and p16: using evolutionary sequence analysis and disease-associated mutations to predict the functional consequences of allelic variants. Oncogene 2003; 22:1150-63. [PMID: 12606942 DOI: 10.1038/sj.onc.1206101] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Deciding whether a missense allelic variant affects protein function is important in many contexts. We previously demonstrated that a detailed analysis of p53 intragenic conservation correlates with somatic mutation hotspots. Here we refine these evolutionary studies and expand them to the p16/Ink4a gene. We calculated that in order for 'absolute conservation' of a codon across multiple species to achieve P<0.05, the evolutionary substitution database must contain at least 3(M) variants, where M equals the number of codons in the gene. Codons in p53 were divided into high (73% of codons), intermediate (29% of codons), and low (0 codons) likelihood of being mutation hotspots. From a database of 263 somatic missense p16 mutations, we identified only four codons that are mutational hotspots at P<0.05 (8 mutations). However, data on function, structure, and disease association support the conclusion that 11 other codons with > or =5 somatic mutations also likely indicate functionally critical residues, even though P0.05. We calculated p16 evolution using amino acid substitution matrices and nucleotide substitution distances. We looked for evolutionary parameters at each codon that would predict whether missense mutations were disease associated or disrupted function. The current p16 evolutionary substitution database is too small to determine whether observations of 'absolute conservation' are statistically significant. Increasing the number of sequences from three to seven significantly improved the predictive value of evolutionary computations. The sensitivity and specificity for conservation scores in predicting disease association of p16 codons is 70-80%. Despite the small p16 sequence database, our calculations of high conservation correctly predicted loss of cell cycle arrest function in 75% of tested codons, and low conservation correctly predicted wild-type function in 80-90% of codons. These data validate our hypothesis that detailed evolutionary analyses help predict the consequences of missense amino-acid variants.
Collapse
Affiliation(s)
- M S Greenblatt
- Department of Medicine, Vermont Cancer Center, University of Vermont, Burlington, VT 05401, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
Rheinwald JG, Hahn WC, Ramsey MR, Wu JY, Guo Z, Tsao H, De Luca M, Catricalà C, O'Toole KM. A two-stage, p16(INK4A)- and p53-dependent keratinocyte senescence mechanism that limits replicative potential independent of telomere status. Mol Cell Biol 2002; 22:5157-72. [PMID: 12077343 PMCID: PMC139780 DOI: 10.1128/mcb.22.14.5157-5172.2002] [Citation(s) in RCA: 259] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
With increasing frequency during serial passage in culture, primary human keratinocytes express p16(INK4A) (p16) and undergo senescence arrest. Keratinocytes engineered to express hTERT maintain long telomeres but typically are not immortalized unless, by mutation or other heritable event, they avoid or greatly reduce p16 expression. We have confirmed that keratinocytes undergo p16-related senescence during growth in culture, whether in the fibroblast feeder cell system or in the specialized K-sfm medium formulation, and that this mechanism can act as a barrier to immortalization following hTERT expression. We have characterized the p16-related arrest mechanism more precisely by interfering specifically with several regulators of cell cycle control. Epidermal, oral mucosal, corneal limbal, and conjunctival keratinocytes were transduced to express a p16-insensitive mutant cdk4 (cdk4(R24C)), to abolish p16 control, and/or a dominant negative mutant p53 (p53DD), to abolish p53 function. Expression of either cdk4(R24C) or p53DD alone had little effect on life span, but expression of both permitted cells to divide 25 to 43 population doublings (PD) beyond their normal limit. Keratinocytes from a p16(+/-) individual transduced to express p53DD alone displayed a 31-PD life span extension associated with selective growth of variants that had lost the wild-type p16 allele. Cells in which both p53 and p16 were nonfunctional divided rapidly during their extended life span but experienced telomere erosion and ultimately ceased growth with very short telomeres. Expression of hTERT in these cells immortalized them. Keratinocytes engineered to express cdk4(R24C) and hTERT but not p53DD did not exhibit an extended life span. Rare immortal variants exhibiting p53 pathway defects arose from them, however, indicating that the p53-dependent component of keratinocyte senescence is telomere independent. Mutational loss of p16 and p53 has been found to be a frequent early event in the development of squamous cell carcinoma. Our results suggest that such mutations endow keratinocytes with extended replicative potential which may serve to increase the probability of neoplastic progression.
Collapse
Affiliation(s)
- James G Rheinwald
- Department of Dermatology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hashemi J, Lindström MS, Asker C, Platz A, Hansson J, Wiman KG. A melanoma-predisposing germline CDKN2A mutation with functional significance for both p16 and p14ARF. Cancer Lett 2002; 180:211-21. [PMID: 12175554 DOI: 10.1016/s0304-3835(02)00027-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The CDKN2A locus on human chromosome 9p21 encodes two proteins, p16 and p14ARF, that mainly regulate cell cycle progression and cell survival via the pRb and p53 pathways, respectively. Germline mutations in CDKN2A have been linked to development of cutaneous melanoma in some families with hereditary melanoma. Due to overlapping open reading frames in exon 2, some mutations in this exon affect both p16 and p14ARF. We previously reported a 24bp deletion in CDKN2A exon 2 in a patient with multiple primary melanomas and melanoma heredity. To further clarify the possible role of the 24bp deletion for melanoma development, especially with respect to p14ARF, we have studied the cellular distribution and function of the resulting p14ARF del (77-84) and p16 del (62-69) mutant proteins. We found that p14ARF del (77-84) had decreased nucleolar localization, and was less efficient than wt p14ARF in stabilizing p53, inducing G1 cell cycle arrest, and inhibiting colony formation. The p16 del (62-69) mutant localized predominantly to the cytoplasm, did not induce G1 cell cycle arrest, and failed to suppress colony formation. We conclude that p14ARF del (77-84) has retained the ability to stabilize MDM2 and p53, but that it is less potent than wt p14ARF. This partial functional defect may complement the clearly defective p16 del (62-69) mutant and thus contribute to melanoma development in patients carrying the 24bp deletion in CDKN2A.
Collapse
Affiliation(s)
- Jamileh Hashemi
- Department of Oncology-Pathology, Research Laboratory of Radiumhemmet, Cancer Center Karolinska, R8:03, Karolinska Hospital, S-171 76 Stockholm, Sweden
| | | | | | | | | | | |
Collapse
|
41
|
Mantelli M, Barile M, Ciotti P, Ghiorzo P, Lantieri F, Pastorino L, Catricalà C, Torre GD, Folco U, Grammatico P, Padovani L, Pasini B, Rovini D, Queirolo P, Rainero ML, Santi PL, Sertoli RM, Goldstein AM, Bianchi-Scarrà G. High prevalence of the G101W germline mutation in theCDKN2A(P16ink4a) gene in 62 Italian malignant melanoma families. ACTA ACUST UNITED AC 2002. [DOI: 10.1002/ajmg.10137] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
42
|
Rizos H, Darmanian AP, Holland EA, Mann GJ, Kefford RF. Mutations in the INK4a/ARF melanoma susceptibility locus functionally impair p14ARF. J Biol Chem 2001; 276:41424-34. [PMID: 11518711 DOI: 10.1074/jbc.m105299200] [Citation(s) in RCA: 82] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The INK4a/ARF locus encodes two cell cycle regulatory proteins, the cyclin-dependent kinase inhibitor, p16(INK4a), and the p53 activator, p14(ARF). Germline mutations in this locus are associated with melanoma susceptibility in 20-40% of multiple case melanoma families. Many of these mutations specifically impair p16(INK4a), whereas mutations uniquely targeting p14(ARF) are rare. Nevertheless, the importance of p14(ARF) has not been excluded because more than 40% of INK4a/ARF alterations affect p16(INK4a) and p14(ARF). We now report that p14(ARF) is functionally impaired in melanoma kindreds carrying INK4a/ARF mutations. Of the seven INK4a/ARF mutations tested, three altered the subcellular distribution of p14(ARF) and diminished the ability of p14(ARF) to activate the p53 pathway. This work establishes the importance of p14(ARF) in melanoma predisposition.
Collapse
Affiliation(s)
- H Rizos
- Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, Westmead Hospital, Westmead, New South Wales 2145, Australia.
| | | | | | | | | |
Collapse
|
43
|
Auroy S, Avril MF, Chompret A, Pham D, Goldstein AM, Bianchi-Scarrà G, Frebourg T, Joly P, Spatz A, Rubino C, Demenais F, Bressac-de Paillerets B. Sporadic multiple primary melanoma cases: CDKN2A germline mutations with a founder effect. Genes Chromosomes Cancer 2001; 32:195-202. [PMID: 11579459 DOI: 10.1002/gcc.1183] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Multiple primary cancers are one of the hallmarks of inherited predisposition. Outside the familial context, multiple primary tumors could be related either to germline de novo mutations or to low-penetrance mutations, in predisposing genes. We selected 100 patients who displayed multiple primary melanoma (MPM) without any known melanoma cases recorded within their families and looked for germline mutations in the two melanoma-predisposing genes identified to date, CDKN2A and CDK4 exon 2. Nine patients (9%) had germline mutations in CDKN2A, whereas none carried germline mutations in exon 2 of CDK4. Seven cases displayed a recurrent missense mutation, G101W, already described in more than 20 melanoma-prone families; one case carried a missense mutation never reported to date (P114S), and the last case was a carrier of a 6 bp insertion at nucleotide 57 resulting in a duplication of codons 18 and 19. To ascertain whether the G101W was a mutational hot spot for de novo mutations or a common founder mutation, we genotyped eight microsatellite markers flanking the CDKN2A gene. After allowing for recombination over time, haplotype sharing provided evidence for an original G101W mutation common to 6 out of 7 sporadic MPM cases. Therefore, it can be concluded that de novo germline CDKN2A mutations associated with MPM are rare.
Collapse
Affiliation(s)
- S Auroy
- Service de Génétique, Institut Gustave Roussy, Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Schraml P, Struckmann K, Bednar R, Fu W, Gasser T, Wilber K, Kononen J, Sauter G, Mihatsch MJ, Moch H. CDKNA2A mutation analysis, protein expression, and deletion mapping of chromosome 9p in conventional clear-cell renal carcinomas: evidence for a second tumor suppressor gene proximal to CDKN2A. THE AMERICAN JOURNAL OF PATHOLOGY 2001; 158:593-601. [PMID: 11159196 PMCID: PMC1850295 DOI: 10.1016/s0002-9440(10)64001-1] [Citation(s) in RCA: 42] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Inactivation of tumor suppressor genes on chromosome 9p is considered a critical event in renal cell carcinoma pathogenesis. Alterations of CDKN2A on 9p21 have been reported in renal cancer cell lines, but their relevance for primary renal carcinomas is unclear. Loss of heterozygosity (LOH) was analyzed by using four polymorphic microsatellites at D9S970 (9p12-9p13), D9S171 (9p13), D9S1748 (9p21), and D9S156 (9p21) in 113 primary conventional clear-cell renal cell carcinomas (CRCCs). Allelic deletion was detected in 21 of 88 informative CRCCs (24%) with the highest rate of LOH being observed at D9S171 on 9p13 (20%). Chromosome 9p LOH was associated with short tumor-specific survival in stage pT3 RCC (P = 0.01). Fluorescence in situ hybridization analysis of 54 CRCCs revealed no homozygous CDKN2A deletions indicating that this mechanism of CDKN2A inactivation is rare in CRCC. Sequencing of 113 CRCCs showed that 13 tumors (12%) had a 24-bp deletion abrogating codons 4 through 11 of CDKN2A. Immunohistochemical CDKN2A expression was absent in normal renal tissue and was only detected in six of 382 CRCCs (1.5%) on a renal tumor microarray. These data suggest that CDKN2A alterations are present in a small subset of CRCCs and a second, yet unknown tumor suppressor gene proximal to the CDKN2A locus, may play a role in CRCC development.
Collapse
Affiliation(s)
- P Schraml
- Institute of Pathology, and the Clinic for Urology, University Basel, Basel, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
In this review, we consider the motivation behind contemporary single nucleotide polymorphism (SNP) initiatives. Many of these initiatives are projected to involve large, population-based surveys. We therefore emphasize the utility of SNPs for genetic epidemiology studies. We start by offering an overview of genetic polymorphism and discuss the historical use of polymorphism in the identification of disease-predisposing genes via meiotic mapping. We next consider some of the unique aspects of SNPs, and their relative advantages and disadvantages in human population-based analyses. In this context, we describe and critique the following six different areas of application for SNP technologies: Gene discovery and mapping. Association-based candidate polymorphism testing. Diagnostics and risk profiling. Prediction of response to environmental stimuli, xenobiotics and diet. Homogeneity testing and epidemiological study design. Physiologic genomics. We focus on key issues within each of these areas in an effort to point out potential problems that might plague the use of SNPs (or other forms of polymorphism) within them. However, we make no claim that our list of considerations are exhaustive. Rather, we believe that they may provide a starting point for further dialog about the ultimate utility of SNP technologies. In addition, although our emphasis is placed on applications of SNPs to the understanding of human phenotypes, we acknowledge that SNP maps and technologies applied to other species (e.g. the mouse genome, pathogen genomes, plant genomes, etc.) are also of tremendous interest.
Collapse
Affiliation(s)
- N J Schork
- Department of Epidemiology and Biostatistics, Case Western Reserve University, Cleveland, OH, USA. @po.cwru.edu
| | | | | |
Collapse
|
46
|
Goldberg EK, Glendening JM, Karanjawala Z, Sridhar A, Walker GJ, Hayward NK, Rice AJ, Kurera D, Tebha Y, Fountain JW. Localization of multiple melanoma tumor-suppressor genes on chromosome 11 by use of homozygosity mapping-of-deletions analysis. Am J Hum Genet 2000; 67:417-31. [PMID: 10877980 PMCID: PMC1287213 DOI: 10.1086/302999] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2000] [Accepted: 05/31/2000] [Indexed: 11/03/2022] Open
Abstract
Loss-of-heterozygosity (LOH) studies have implicated one or more chromosome 11 tumor-suppressor gene(s) in the development of cutaneous melanoma as well as a variety of other forms of human cancer. In the present study, we have identified multiple independent critical regions on this chromosome by use of homozygosity mapping of deletions (HOMOD) analysis. This method of analysis involved the use of highly polymorphic microsatellite markers and statistics to identify regions of hemizygous deletion in unmatched melanoma cell line DNAs. Regions of loss were defined by the presence of an extended region of homozygosity (ERH) at > or =5 adjacent markers and having a statistical probability of < or =.001. Significant ERHs were similar in nature to deletions identified by LOH analyses performed on uncultured melanomas, although a higher frequency of loss (24 [60%] of 40 vs. 16 [34%] of 47) was observed in the cell lines. Overall, six small regions of overlapping deletions (SROs) were identified on chromosome 11 flanked by the markers D11S1338/D11S907 (11p13-15.5 [SRO1]), D11S1344/D11S11385 (11p11.2 [SRO2]), D11S917/D11S1886 (11q21-22.3 [SRO3]), D11S927/D11S4094 (11q23 [SRO4]), AFM210ve3/D11S990 (11q24 [SRO5]), and D11S1351/D11S4123 (11q24-25 [SRO6]). We propose that HOMOD analysis can be used as an adjunct to LOH analysis in the localization of tumor-suppressor genes.
Collapse
Affiliation(s)
- Eleonora K. Goldberg
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - J. Michael Glendening
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Zarir Karanjawala
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Anjali Sridhar
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Graeme J. Walker
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Nicholas K. Hayward
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Andrew J. Rice
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Devinda Kurera
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Yasmine Tebha
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | - Jane W. Fountain
- Institute for Genetic Medicine, Department of Biochemistry and Molecular Biology, University of Southern California, Los Angeles; and Queensland Cancer Fund Research Unit, Joint Experimental Oncology Program of the Queensland Institute of Medical Research and the University of Queensland, Royal Brisbane Hospital, Herston, Australia
| |
Collapse
|