1
|
Bae SY, Ling HH, Chen Y, Chen H, Kumar D, Zhang J, Viny AD, DePinho RA, Giancotti FG. Mediator Subunit Med4 Enforces Metastatic Dormancy in Breast Cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.11.18.566087. [PMID: 38014033 PMCID: PMC10680920 DOI: 10.1101/2023.11.18.566087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2023]
Abstract
Long term survival of breast cancer patients is limited due to recurrence from metastatic dormant cancer cells. However, the mechanisms by which these dormant breast cancer cells survive and awaken remain poorly understood. Our unbiased genome-scale genetic screen in mice identified Med4 as a novel cancer-cell intrinsic gatekeeper in metastatic reactivation. MED4 haploinsufficiency is prevalent in metastatic breast cancer patients and correlates with poorer prognosis. Syngeneic xenograft models revealed that Med4 enforces breast cancer dormancy. Contrary to the canonical function of the Mediator complex in activating gene expression, Med4 maintains 3D chromatin compaction and enhancer landscape, by preventing enhancer priming or activation through the suppression of H3K4me1 deposition. Med4 haploinsufficiency disrupts enhancer poise and reprograms the enhancer dynamics to facilitate extracellular matrix (ECM) gene expression and integrin-mediated mechano-transduction, driving metastatic growth. Our findings establish Med4 as a key regulator of cellular dormancy and a potential biomarker for high-risk metastatic relapse.
Collapse
Affiliation(s)
- Seong-Yeon Bae
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Hsiang-Hsi Ling
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Yi Chen
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Columbia Stem Cell Initiative, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Hong Chen
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Department of Thoracic/Head and Neck Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Dhiraj Kumar
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Jiankang Zhang
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Aaron D. Viny
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
- Columbia Stem Cell Initiative, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Division of Hematology and Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, New York 10032, USA
| | - Ronald A. DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
| | - Filippo G. Giancotti
- Cancer Metastasis Initiative, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Genetics and Development, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York 10032, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas 77054, USA
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, New York 10032, USA
| |
Collapse
|
2
|
Price EA, Sagoo MS, Reddy MA, Onadim Z. An overview of RB1 transcript alterations detected during retinoblastoma genetic screening. Ophthalmic Genet 2024; 45:235-245. [PMID: 37932244 DOI: 10.1080/13816810.2023.2270570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/24/2023] [Accepted: 10/09/2023] [Indexed: 11/08/2023]
Abstract
Identification of pathogenic RB1 variants aids in the clinical management of families with retinoblastoma. We routinely screen DNA for RB1 variants, but transcript analysis can also be used for variant screening, and to help decide variant pathogenicity. DNA was screened by conformation analysis followed by Sanger sequencing. Large deletion/insertions were detected by polymorphism analysis, MLPA and quantitative-PCR. Methylation-specific PCR was used to detect hypermethylation. RNA screening was performed when a DNA pathogenic variant was missing, or to determine effects on splicing.Two hundred and thirteen small coding variants were predicted to affect splicing in 207 patients. Splice donor (sd) variants were nearly twice as frequent as splice acceptor (sa) with the most affected positions being sd + 1 and sa-1. Some missense and nonsense codons altered splicing, while some splice consensus variants did not. Large deletion/insertions can disrupt splicing, but RNA analysis showed that some of these are more complex than indicated by DNA testing. RNA screening found pathogenic variants in 53.8% of samples where DNA analysis did not. RB1 splicing is altered by changes at consensus splice sites, some missense and nonsense codons, deep intronic changes and large deletion/insertions. Common alternatively spliced transcripts may complicate analysis. An effective molecular screening strategy would include RNA analysis to help determine pathogenicity.
Collapse
Affiliation(s)
- Elizabeth A Price
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
| | - Mandeep S Sagoo
- Retinoblastoma Service, Royal London Hospital, Barts Health NHS Trust, London, UK
- NIHR Biomedical Research Centre for Ophthalmology, Moorfields Eye Hospital, Institute of Ophthalmology, University College London, London, UK
| | - M Ashwin Reddy
- Retinoblastoma Service, Royal London Hospital, Barts Health NHS Trust, London, UK
- Faculty of Medicine, Queen Mary University of London, London, UK
| | - Zerrin Onadim
- Retinoblastoma Genetic Screening Unit, Barts Health NHS Trust, London, UK
- Faculty of Medicine, Queen Mary University of London, London, UK
| |
Collapse
|
3
|
Prenatal diagnosis of distal 13q deletion syndrome in a fetus with esophageal atresia: a case report and review of the literature. J Med Case Rep 2022; 16:481. [PMID: 36572904 PMCID: PMC9793530 DOI: 10.1186/s13256-022-03713-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Accepted: 12/07/2022] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Chromosome 13q deletion syndrome shows variable clinical features related to the different potential breakpoints in chromosome 13q. The severely malformed phenotype is known to be associated with the deletion of a critical region in 13q32. However, esophageal atresia is a rare symptom and the relevant region is unknown. Thus, determining the association between accurate breakpoints and new clinical features is essential. CASE PRESENTATION A 28-year-old Japanese primigravid woman was referred for fetal growth restriction, absence of a gastric bubble, cerebellar hypoplasia, overlapping fingers, and polyhydramnios at 31 weeks gestation. At 38 + 0 weeks, she delivered a 1774 g female infant. The infant presented with isolated esophageal atresia (Gross type A), Dandy-Walker malformation, right microphthalmia, left coloboma, overlapping fingers, pleurocentrum in the thoracic vertebrae, reduced anogenital distance, and hearing loss. Her karyotype was diagnosed as 46,XX,del(13)(q32.1-qter) by amniocentesis, but array comparative genomic hybridization after birth revealed the deletion of 13q31.3-qter. At 48 days after birth, the infant underwent surgery for esophageal atresia and was later discharged from the hospital at 7 months of age. CONCLUSION This case report and the literature reviews supports the previous findings on the pathological roles of haploinsufficiency of the ZIC2/ZIC5 in Dandy-Walker malformation and the EFBN2 haploinsufficiency in eye malformation and hearing loss. Furthermore, the possible involvement of IRS2, COLA1, and COLA2 in eye malformation were identified. This is the first case of 13q deletion syndrome with esophageal atresia (Gross A), but it may be a symptom of VATER/VACTER association (vertebral defects, anorectal malformations, cardiac defects, tracheoesophageal fistula with or without esophageal atresia, renal malformations, and limb defects), as in the previous cases. These symptoms might also be associated with EFBN2 haploinsufficiency, although further research is required.
Collapse
|
4
|
Mendonça V, Pereira Sena P, Evangelista Dos Santos AC, Rodrigues Bonvicino C, Ashton-Prolla P, Epelman S, Ferman SE, Lapunzina P, Nevado J, Grigorovski N, Mattosinho C, Seuànez H, Regla Vargas F. Diverse mutational spectrum in the 13q14 chromosomal region in a Brazilian cohort of retinoblastoma. Exp Eye Res 2022; 224:109211. [PMID: 35985532 DOI: 10.1016/j.exer.2022.109211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/19/2022] [Accepted: 08/03/2022] [Indexed: 11/27/2022]
Abstract
Retinoblastoma is a rare childhood tumor caused by the inactivation of both copies of the RB1 gene. Early diagnosis and identification of heritable RB1 mutation carriers can improve the disease outcome and management via genetic counseling. We used the Multiplex Ligation-dependent Probe Amplification (MLPA) method to analyze the RB1 gene and flanking regions in blood samples from 159 retinoblastoma patients previously negative for RB1 point mutations via Sanger sequencing. We detected a wide spectrum of germline chromosomal alterations, ranging from partial loss or duplication of RB1 to large deletions spanning RB1 and adjacent genes. Mutations were validated via karyotyping, fluorescent in situ hybridization (FISH), SNP-arrays (Single Nucleotide Polymorphism-arrays) and/or quantitative relative real-time PCR. Patients with leukocoria as a presenting symptom showed reduced death rate (p = 0.013) and this sign occurred more frequently among carriers of two breakpoints within RB1 (p = 0.05). All unilateral cases presented both breakpoints outside of RB1 (p = 0.0075). Patients with one breakpoint within RB1 were diagnosed at earlier ages (p = 0.017). Our findings characterize the mutational spectrum of a Brazilian cohort of retinoblastoma patients and point to a possible relationship between the mutation breakpoint location and tumor outcome, contributing to a better prospect of the genotype/phenotype correlation and adding to the wide diversity of germline mutations involving RB1 and adjacent regions in retinoblastoma.
Collapse
Affiliation(s)
- Vanessa Mendonça
- Genetics Program, Instituto Nacional de Câncer, Rio de Janeiro, Brazil; Genetics Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Priscila Pereira Sena
- Genetics Program, Instituto Nacional de Câncer, Rio de Janeiro, Brazil; Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | | | | | | | - Sidnei Epelman
- Pediatric Oncology Service, Hospital Santa Marcelina, São Paulo, Brazil
| | - Sima Esther Ferman
- Department of Pediatric Oncology, Clinical Division, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Pablo Lapunzina
- INGEMM, Hospital La Paz, Universidad de Madrid, Madrid, Spain; CIBERER (Centro de Investigación Médica en Red de Enfermedades Raras), Madrid, Spain; ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Julián Nevado
- INGEMM, Hospital La Paz, Universidad de Madrid, Madrid, Spain; CIBERER (Centro de Investigación Médica en Red de Enfermedades Raras), Madrid, Spain; ITHACA-European Reference Network, Hospital La Paz, Madrid, Spain
| | - Nathalia Grigorovski
- Department of Pediatric Oncology, Clinical Division, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Clarissa Mattosinho
- Department of Ocular Oncology, Division of Surgery, Instituto Nacional de Câncer, Rio de Janeiro, Brazil
| | - Hector Seuànez
- Genetics Program, Instituto Nacional de Câncer, Rio de Janeiro, Brazil; Genetics Department, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Fernando Regla Vargas
- Birth Defects Epidemiology Laboratory, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brazil; Department of Genetics and Molecular Biology, Universidade Federal do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
5
|
Tunç E, Ilgaz S. Robertsonian translocation (13;14) and its clinical manifestations: A literature review. Reprod Biomed Online 2022; 45:563-573. [DOI: 10.1016/j.rbmo.2022.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 04/06/2022] [Accepted: 05/25/2022] [Indexed: 10/18/2022]
|
6
|
Watson CM, Holliday DL, Crinnion LA, Bonthron DT. Long‐read nanopore DNA sequencing can resolve complex intragenic duplication/deletion variants, providing information to enable preimplantation genetic diagnosis. Prenat Diagn 2022; 42:226-232. [PMID: 35014072 PMCID: PMC9305782 DOI: 10.1002/pd.6089] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 11/15/2021] [Accepted: 12/31/2021] [Indexed: 11/11/2022]
Abstract
Background Objective Methods Results Conclusion
What's already known about this topic?
Molecular diagnostic techniques that incompletely resolve pathogenic sequence variants can present a barrier for certain prenatal diagnostic approaches.
What does this study add?
This study demonstrates how nanopore‐based sequencing could be rapidly deployed for follow‐up analysis of previously identified, but incompletely‐defined structural variants, enabling onward referral to a national preimplantation genetic diagnosis service.
Collapse
Affiliation(s)
- Christopher M. Watson
- North East and Yorkshire Genomics Laboratory Hub Central Laboratory St. James's University Hospital Leeds UK
- Leeds Institute of Medical Research University of Leeds St. James's University Hospital Leeds UK
| | - Deborah L. Holliday
- Department of Clinical Genetics Leeds Teaching Hospitals NHS Trust Chapel Allerton Hospital Leeds West Yorkshire UK
| | - Laura A. Crinnion
- North East and Yorkshire Genomics Laboratory Hub Central Laboratory St. James's University Hospital Leeds UK
- Leeds Institute of Medical Research University of Leeds St. James's University Hospital Leeds UK
| | - David T. Bonthron
- Leeds Institute of Medical Research University of Leeds St. James's University Hospital Leeds UK
- Department of Clinical Genetics Leeds Teaching Hospitals NHS Trust Chapel Allerton Hospital Leeds West Yorkshire UK
| |
Collapse
|
7
|
Keuper K, Wieland A, Räschle M, Storchova Z. Processes shaping cancer genomes - From mitotic defects to chromosomal rearrangements. DNA Repair (Amst) 2021; 107:103207. [PMID: 34425515 DOI: 10.1016/j.dnarep.2021.103207] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 08/06/2021] [Accepted: 08/07/2021] [Indexed: 11/19/2022]
Abstract
Sequencing of cancer genomes revealed a rich landscape of somatic single nucleotide variants, structural changes of chromosomes, as well as chromosomal copy number alterations. These chromosome changes are highly variable, and simple translocations, deletions or duplications have been identified, as well as complex events that likely arise through activity of several interconnected processes. Comparison of the cancer genome sequencing data with our knowledge about processes important for maintenance of genome stability, namely DNA replication, repair and chromosome segregation, provides insights into the mechanisms that may give rise to complex chromosomal patterns, such as chromothripsis, a complex form of multiple focal chromosome rearrangements. In addition, observations gained from model systems that recapitulate the rearrangements patterns under defined experimental conditions suggest that mitotic errors and defective DNA replication and repair contribute to their formation. Here, we review the molecular mechanisms that contribute to formation of chromosomal aberrations observed in cancer genomes.
Collapse
Affiliation(s)
- Kristina Keuper
- Department of Molecular Genetics, Paul-Ehrlich Strasse 24, University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Angela Wieland
- Department of Molecular Genetics, Paul-Ehrlich Strasse 24, University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Markus Räschle
- Department of Molecular Genetics, Paul-Ehrlich Strasse 24, University of Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Zuzana Storchova
- Department of Molecular Genetics, Paul-Ehrlich Strasse 24, University of Kaiserslautern, 67663, Kaiserslautern, Germany.
| |
Collapse
|
8
|
Gargallo P, Oltra S, Balaguer J, Barranco H, Yáñez Y, Segura V, Juan-Ribelles A, Calabria I, Llavador M, Castel V, Cañete A. Retinoblastoma and mosaic 13q deletion: a case report. Int J Retina Vitreous 2021; 7:50. [PMID: 34479642 PMCID: PMC8418035 DOI: 10.1186/s40942-021-00321-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Patients with 13q-syndrome are at risk of retinoblastoma when the RB1 gene, located in the chromosomal band 13q14.2, is deleted. This syndrome is frequently associated with congenital malformations and developmental delay, although these signs could be mild. Mosaic 13q-deletion patients have been previously reported in the literature; their phenotype is variable, and they may not be recognized. CASE PRESENTATION Retinoblastoma diagnosed in a child with 13q-mosaicism confirmed in blood, oral mucosa, healthy retina and retinoblastoma. A second RB1 hit is present exclusively in the retinoblastoma sample (RB1 c.958C>T p.Arg320Ter). Other detected molecular events in retinoblastoma are 6p12.3pter gain and 6q25.3qter loss. Clinical examination is unremarkable except for clinodactyly of the right fifth finger. DISCUSSION AND CONCLUSIONS We describe a case of mosaic 13q deletion syndrome affected by retinoblastoma. Molecular data obtained from the tumor analysis are similar to previous data available about this malignancy. High clinical suspicion is essential for an adequate diagnosis of mosaic cases.
Collapse
Affiliation(s)
- Pablo Gargallo
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain. .,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.
| | - Silvestre Oltra
- Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain
| | - Julia Balaguer
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain.,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.,Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain.,Pediatric Oncology and Hematology Unit, University Hospital La Fe, Valencia, Spain
| | - Honorio Barranco
- Pediatric Ophthalmology Unit, University Hospital La Fe Hospital, Valencia, Spain
| | - Yania Yáñez
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain
| | - Vanessa Segura
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain
| | - Antonio Juan-Ribelles
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain.,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.,Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain.,Pediatric Oncology and Hematology Unit, University Hospital La Fe, Valencia, Spain
| | - Inés Calabria
- Oncology Department, Imegen-Health in Code Group, Valencia, Spain
| | | | - Victoria Castel
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain
| | - Adela Cañete
- Clinical and Translational Research in Cancer, Health Research Institute La Fe (IIS La Fe), Valencia, Spain.,Oncology Department, Imegen-Health in Code Group, Valencia, Spain.,Genetics Unit, La Fe Hospital, Valencia, Spain.,Genetics Department, Valencia University, Valencia, Spain.,Pediatric Oncology and Hematology Unit, University Hospital La Fe, Valencia, Spain
| |
Collapse
|
9
|
|
10
|
Privitera F, Calonaci A, Doddato G, Papa FT, Baldassarri M, Pinto AM, Mari F, Longo I, Caini M, Galimberti D, Hadjistilianou T, De Francesco S, Renieri A, Ariani F. 13q Deletion Syndrome Involving RB1: Characterization of a New Minimal Critical Region for Psychomotor Delay. Genes (Basel) 2021; 12:1318. [PMID: 34573300 PMCID: PMC8471443 DOI: 10.3390/genes12091318] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 08/24/2021] [Accepted: 08/24/2021] [Indexed: 11/17/2022] Open
Abstract
Retinoblastoma (RB) is an ocular tumor of the pediatric age caused by biallelic inactivation of the RB1 gene (13q14). About 10% of cases are due to gross-sized molecular deletions. The deletions can involve the surrounding genes delineating a contiguous gene syndrome characterized by RB, developmental anomalies, and peculiar facial dysmorphisms. Overlapping deletions previously found by traditional and/or molecular cytogenetic analysis allowed to define some critical regions for intellectual disability (ID) and multiple congenital anomalies, with key candidate genes. In the present study, using array-CGH, we characterized seven new patients with interstitial 13q deletion involving RB1. Among these cases, three patients with medium or large 13q deletions did not present psychomotor delay. This allowed defining a minimal critical region for ID that excludes the previously suggested candidate genes (HTR2A, NUFIP1, PCDH8, and PCDH17). The region contains 36 genes including NBEA, which emerged as the candidate gene associated with developmental delay. In addition, MAB21L1, DCLK1, EXOSC8, and SPART haploinsufficiency might contribute to the observed impaired neurodevelopmental phenotype. In conclusion, this study adds important novelties to the 13q deletion syndrome, although further studies are needed to better characterize the contribution of different genes and to understand how the haploinsufficiency of this region can determine ID.
Collapse
Affiliation(s)
- Flavia Privitera
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Arianna Calonaci
- Unit of Pediatrics, Department of Maternal, Newborn and Child Health, Azienda Ospedaliera Universitaria Senese, Policlinico ‘Santa Maria alle Scotte’, 53100 Siena, Italy; (A.C.); (M.C.); (D.G.)
| | - Gabriella Doddato
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Filomena Tiziana Papa
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Margherita Baldassarri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
| | - Anna Maria Pinto
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (A.M.P.); (I.L.)
| | - Francesca Mari
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (A.M.P.); (I.L.)
| | - Ilaria Longo
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (A.M.P.); (I.L.)
| | - Mauro Caini
- Unit of Pediatrics, Department of Maternal, Newborn and Child Health, Azienda Ospedaliera Universitaria Senese, Policlinico ‘Santa Maria alle Scotte’, 53100 Siena, Italy; (A.C.); (M.C.); (D.G.)
| | - Daniela Galimberti
- Unit of Pediatrics, Department of Maternal, Newborn and Child Health, Azienda Ospedaliera Universitaria Senese, Policlinico ‘Santa Maria alle Scotte’, 53100 Siena, Italy; (A.C.); (M.C.); (D.G.)
| | - Theodora Hadjistilianou
- Unit of Ophthalmology and Retinoblastoma Referral Center, Department of Surgery, University of Siena, Policlinico ‘Santa Maria alle Scotte’, 53100 Siena, Italy; (T.H.); (S.D.F.)
| | - Sonia De Francesco
- Unit of Ophthalmology and Retinoblastoma Referral Center, Department of Surgery, University of Siena, Policlinico ‘Santa Maria alle Scotte’, 53100 Siena, Italy; (T.H.); (S.D.F.)
| | - Alessandra Renieri
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (A.M.P.); (I.L.)
| | - Francesca Ariani
- Medical Genetics, University of Siena, 53100 Siena, Italy; (F.P.); (G.D.); (F.T.P.); (M.B.); (F.M.); (A.R.)
- Med Biotech Hub and Competence Center, Department of Medical Biotechnologies, University of Siena, 53100 Siena, Italy
- Genetica Medica, Azienda Ospedaliera Universitaria Senese, 53100 Siena, Italy; (A.M.P.); (I.L.)
| |
Collapse
|
11
|
Shen A, Wu M, Liu L, Chen Y, Chen X, Zhuang M, Xie Q, Cheng Y, Li J, Shen Z, Wei L, Chu J, Sferra TJ, Zhang X, Xu N, Li L, Peng J, Chen F. Targeting NUFIP1 Suppresses Growth and Induces Senescence of Colorectal Cancer Cells. Front Oncol 2021; 11:681425. [PMID: 34367967 PMCID: PMC8343530 DOI: 10.3389/fonc.2021.681425] [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] [Received: 03/16/2021] [Accepted: 07/05/2021] [Indexed: 12/01/2022] Open
Abstract
NUFIP1 is an RNA-binding protein that interacts with fragile X mental retardation protein (FMRP) in the messenger ribonucleoprotein particle (mRNP). We previously showed that NUFIP1 was upregulated in colorectal cancer (CRC), but how the protein may contribute to the disease and patient prognosis is unknown. Here we combine database analysis, microarray, quantitative PCR, and immunohistochemistry of patients' samples to confirm our previous findings on NUFIP1 overexpression in CRC, and to reveal that increased expression of NUFIP1 in CRC tissues correlated with worse overall, recurrence-free, event-free and disease-free survival in patients, as well as with more advanced CRC clinicopathological stage. Loss of function analysis demonstrated that NUFIP1 knockdown suppressed cell growth in vitro and in vivo, inhibited cell viability and survival, and induced cell cycle arrest and apoptosis in vitro, as well as up-regulated Bax and down-regulated Bcl-2 protein expression. In addition, as a natural anticancer triterpene from various fruits and vegetables, ursolic acid (UA) treatment suppressed cell proliferation, down-regulated NUFIP1 protein expression, and further enhanced the effects of NUFIP1 knockdown in CRC cells in vitro. NUFIP1 knockdown up-regulated the expression of 136 proteins, down-regulated the expression of 41 proteins, and enriched multiple signaling pathways including the senescence-associated heterochromatin foci (SAHF) pathway. Furthermore, NUFIP1 knockdown enhanced the expression of senescence-associated-β-galactosidase (SA-β-gal), the SAHF markers HP1γ and trimethylation (H3k9me3), and the senescence-related protein HMGA2, as well as both p53 and its downstream p21 protein expression. Our findings suggest that NUFIP1 is overexpressed in CRC and correlates with disease progression and poor patient survival. NUFIP1 may exert oncogenic effects partly by altering senescence. UA may show potential to treat CRC by down-regulating NUFIP1.
Collapse
Affiliation(s)
- Aling Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Meizhu Wu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Liya Liu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Youqin Chen
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children’s Hospital, Cleveland, OH, United States
| | - Xiaoping Chen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Mingkai Zhuang
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Qiurong Xie
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Ying Cheng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jiapeng Li
- Department of Physical Education, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Zhiqing Shen
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lihui Wei
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Jianfeng Chu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Thomas J. Sferra
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children’s Hospital, Cleveland, OH, United States
| | - Xiuli Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Nanhui Xu
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Li Li
- Department of Health Management, Fujian Provincial Hospital, Shengli Clinical College of Fujian Medical University, Fuzhou, China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Fenglin Chen
- Department of Gastroenterology, Fujian Medical University Union Hospital, Fuzhou, China
| |
Collapse
|
12
|
Dangles MT, Davous D, Vialle G, Auvrignon A, Angellier E, Bourdeaut F. [Intellectual disability and cancer in children: An analysis of the decision-making process]. Bull Cancer 2021; 108:813-826. [PMID: 34176585 DOI: 10.1016/j.bulcan.2021.02.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/21/2021] [Accepted: 02/26/2021] [Indexed: 11/25/2022]
Abstract
AIM The aim was to describe and to analyze the ethics of decision-making in situations involving children with intellectual disability and cancer, from the referent-doctor's point-of-view, in pediatric oncology units in France. METHODS Pediatricians working in pediatric oncology units were interviewed through an online questionnaire and a semi-directive interview was systematically proposed. We analyzed the ethical issues that arose during the process of decision-making and we made suggestions in order to address them. RESULTS Sixteen doctors reported twenty-one clinical cases. Of these cases, one third of the children had a change in their oncologic treatment, with a risk of pejorative outcome on the prognosis. Despite the fact that ethical issues appeared in 80 % of the cases, there were few consultations with ethical committees. Decision-making process showed no difference compared to children without intellectual disability, thus raising ethical issues in the medical team. Our study showed discrepancy between frequently reported ethical issues, high consensus rate regarding treatment decision and lack of consultation with ethical committees. DISCUSSION We propose three steps to guide the decision-making process in situations involving children with intellectual disability and cancer: 1/deeper understanding of the child through reinforced interactions with their caregivers, 2/better cross-boundary discussions, to improve the effectiveness of the multidisciplinary staff, and 3/more systematic ethical committees consultation.
Collapse
Affiliation(s)
- Marie-Thérèse Dangles
- Hôpital Necker-Enfants-Malades, service de neurologie pédiatrique, 149, rue de Sèvres, 75015 Paris, France; Université de Paris, Paris, France.
| | - Dominique Davous
- Hôpital Saint-Louis, groupe de réflexion et de recherche au sein de l'espace éthique région Île-de-France : parents et soignants face à l'éthique en pédiatrie, 1, avenue Claude-Vellefaux, 75010 Paris, France
| | - Guénola Vialle
- Chargée de mission PALIPED-RIFHOP, réseau d'Île-de-France pour l'hématologie, l'oncologie et les soins palliatifs pédiatrique, 3-5, rue de Metz, 75010 Paris, France
| | - Anne Auvrignon
- Hôpital Armand-Trousseau, service d'hématologie pédiatrique, 26, avenue du Dr Arnold-Netter, 75012 Paris, France
| | - Elisabeth Angellier
- Institut Curie, département interdisciplinaire de soins de support pour le patient en oncologie (DISSPO), 35, rue Dailly, 92210 Saint-Cloud, France
| | - Franck Bourdeaut
- Institut Curie, service d'oncologie pédiatrique, SIREDO, 26, rue d'Ulm, 75005 Paris, France
| |
Collapse
|
13
|
Cobbs LV, Francis JH, Dunkel IJ, Gobin YP, Brodie SE, Abramson DH. Retinoblastoma management in 13q deletion syndrome patients using super-selective chemotherapies and other cancer-directed interventions. Pediatr Blood Cancer 2021; 68:e28845. [PMID: 33355996 PMCID: PMC9316827 DOI: 10.1002/pbc.28845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND This study aimed to identify best practices for treating 13q deletion syndrome (13q-) patients with retinoblastoma in the era of super-selective ophthalmic artery chemosurgery (OAC) and intravitreal injection therapy (IVIT). METHODS Retrospective study of 21 eyes from 14 patients with retinoblastoma and 13q- who were treated at Memorial Sloan Kettering Cancer Center (MSKCC) between May 2006 and May 2020, with a mean follow up of 3.7 years. Ocular survival, patient survival, and treatment toxicities were assessed. RESULTS Nine of the 12 eyes that underwent OAC/IVIT at MSKCC have been progression free for at least 1 year since their last treatments. Fifteen out of 26 OAC cycles resulted in grade 3-4 hematologic toxicity. There was one death from sepsis in the setting of intravenous chemotherapy (IVC) for metastatic disease that occurred after OAC/IVIT therapy. The 2-year Kaplan-Meier ocular survival estimate for the whole cohort was 75% and for the eyes that received OAC or IVIT at MSKCC 83%. For OAC hematologic toxicities, one platelet transfusion and two filgrastim doses were administered, and one patient was hospitalized for neutropenic fevers. CONCLUSIONS The majority of 13q- eyes treated with OAC/IVIT-based regimens can be cured, and there were no deaths related to complications from OAC or IVIT. 13q- Patients did have increased risk of systemic treatment complications, even from super-selective chemotherapies. Despite these toxicities, only one patient developed febrile neutropenia, one patient required a blood product transfusion, and two patients received filgrastim for both OAC and IVC complications. PRÉCIS: Children with 13q deletion syndrome with retinoblastoma managed with intra-arterial and intravitreal chemotherapy have excellent patient and ocular survival with acceptable toxicity.
Collapse
Affiliation(s)
- Lucy V. Cobbs
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Jasmine H. Francis
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Ophthalmology, Weill Cornell Medical College, New York Presbyterian Hospital, New York, New York
| | - Ira J. Dunkel
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Pediatrics, Weill Cornell Medical College, New York Presbyterian Hospital, New York, New York
| | - Y Pierre Gobin
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York,Interventional Neuroradiology, Department of Neurosurgery, Weill Cornell Medicine, New York Presbyterian Hospital, New York, New York
| | - Scott E. Brodie
- Department of Ophthalmology, NYU Langone Health, New York, New York
| | - David H. Abramson
- Ophthalmic Oncology Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York,Department of Ophthalmology, Weill Cornell Medical College, New York Presbyterian Hospital, New York, New York
| |
Collapse
|
14
|
Hülsenbeck I, Frank M, Biewald E, Kanber D, Lohmann DR, Ketteler P. Introduction of a Variant Classification System for Analysis of Genotype-Phenotype Relationships in Heritable Retinoblastoma. Cancers (Basel) 2021; 13:cancers13071605. [PMID: 33807189 PMCID: PMC8037437 DOI: 10.3390/cancers13071605] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Revised: 03/26/2021] [Accepted: 03/28/2021] [Indexed: 11/22/2022] Open
Abstract
Simple Summary Heritable retinoblastoma is a genetic disease that predisposes to develop multiple retinoblastomas in childhood and other extraocular tumors later in life. It is caused by genetic variants in the RB1 gene. Here we present a new classification for genetic variants in the RB1 gene (REC) that focuses on the variant’s effect. The different classes, REC-I to -V, correlate with different risks of tumor predisposition. REC correlated with different clinical courses when applied in our study cohort. REC aims to facilitate risk estimation for physicians, patients and their families, and researchers and to improve the definition of the necessity of screening examination. Abstract Constitutional haploinsufficiency of the RB1 gene causes heritable retinoblastoma, a tumor predisposition syndrome. Patients with heritable retinoblastoma develop multiple retinoblastomas early in childhood and other extraocular tumors later in life. Constitutional pathogenic variants in RB1 are heterogeneous, and a few genotype-phenotype correlations have been described. To identify further genotype-phenotype relationships, we developed the retinoblastoma variant effect classification (REC), which considers each variant’s predicted effects on the common causal mediator, RB1 protein pRB. For validation, the RB1 variants of 287 patients were grouped according to REC. Multiple aspects of phenotypic expression were analyzed, known genotype-phenotype associations were revised, and new relationships were explored. Phenotypic expression of patients with REC-I, -II, and -III was distinct. Remarkably, the phenotype of patients with variants causing residual amounts of truncated pRB (REC-I) was more severe than patients with complete loss of RB1 (REC-II). The age of diagnosis of REC-I variants appeared to be distinct depending on truncation’s localization relative to pRB structure domains. REC classes identify genotype-phenotype relationships and, therefore, this classification framework may serve as a tool to develop tailored tumor screening programs depending on the type of RB1 variant.
Collapse
Affiliation(s)
- Isabel Hülsenbeck
- Department of Pediatric Hematology and Oncology, University Duisburg-Essen, University Hospital Essen, Hufelandstrasse 55, 45122 Essen, Germany;
- Eye Oncogenetics Research Group, University Hospital Essen, 45122 Essen, Germany; (D.K.); (D.R.L.)
| | - Mirjam Frank
- Institute for Medical Informatics, Biometry and Epidemiology, University Duisburg-Essen, University Hospital Essen, 45122 Essen, Germany;
| | - Eva Biewald
- Department of Ophthalmology, University Duisburg-Essen, University Hospital Essen, 45122 Essen, Germany;
| | - Deniz Kanber
- Eye Oncogenetics Research Group, University Hospital Essen, 45122 Essen, Germany; (D.K.); (D.R.L.)
- Institute of Human Genetics, University Duisburg-Essen, 45122 Essen, Germany
| | - Dietmar R. Lohmann
- Eye Oncogenetics Research Group, University Hospital Essen, 45122 Essen, Germany; (D.K.); (D.R.L.)
- Institute of Human Genetics, University Duisburg-Essen, 45122 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, 69120 Heidelberg, Germany
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Duisburg-Essen, University Hospital Essen, Hufelandstrasse 55, 45122 Essen, Germany;
- Eye Oncogenetics Research Group, University Hospital Essen, 45122 Essen, Germany; (D.K.); (D.R.L.)
- Institute of Human Genetics, University Duisburg-Essen, 45122 Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, 69120 Heidelberg, Germany
- Correspondence:
| |
Collapse
|
15
|
Abstract
Retinoblastoma is the most common ocular malignancy of childhood. With an estimated 300 cases annually in the United States, retinoblastoma is nevertheless considered a rare tumor. Although retinoblastoma primarily affects younger children, diagnosis during the neonatal age range is less common. However, an understanding of patients at risk is critical for appropriate screening. Early detection and treatment by a multidisciplinary specialty team maximizes the chance for survival and ocular/vision salvage while minimizing treatment-related toxicity. Testing for alterations in the RB1 gene has become standard practice, and informs screening and genetic counseling recommendations for patients and their families.
Collapse
|
16
|
Zhang W, Lei T, Fu F, Deng Q, Li R, Wang D, Yang X, Li D, Liao C. Microarray analysis in fetuses with duodenal obstruction: It is not just trisomy 21. Prenat Diagn 2021; 41:316-322. [PMID: 33000500 DOI: 10.1002/pd.5834] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Revised: 08/10/2020] [Accepted: 09/29/2020] [Indexed: 12/28/2022]
Abstract
OBJECTIVE To explore the copy number variants (CNVs) in case of fetal duodenal obstruction (DO) and assess the associated prenatal findings and postnatal outcomes. MATERIALS AND METHODS This retrospective study reviewed 51 fetuses with DO and the findings of chromosomal microarray analysis (CMA) used as a first-tier test in our institution between January 2014 and May 2019. RESULTS The frequency of pathogenic aberrations in fetuses with DO was 15.7% (8/51), including 9.8% (5/51) pathogenic CNVs. Three fetuses with isolated DO each had a deletion on chromosome 13q, one fetus had duplication at 1q43q44, and one had microduplication at 17q12. No significant differences in pathogenic CNVs were observed between isolated DO and DO plus additional anomalies (4/42, 9.5% vs 1/9, 11.1%, P = .89). Of the 51 fetuses with DO, 11 pregnancies were terminated, and eight fetuses had chromosomal abnormalities; one pregnancy ended with intrauterine death, and there were 39 live births. Neonatal outcomes were available for 31 fetuses, and no neonatal deaths occurred after surgery. CONCLUSIONS Our cohort study demonstrated the value of CMA in fetuses with DO, suggesting that CNVs may underly genetic etiologies that should be considered in the diagnostic evaluation of DO. We think CMA should be recommended in case of DO.
Collapse
Affiliation(s)
- Wenwen Zhang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Tingying Lei
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Fang Fu
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Qiong Deng
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ru Li
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Dan Wang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xin Yang
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Dongzhi Li
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Can Liao
- Department of Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
17
|
Reddy MA, Butt M, Hinds AM, Duncan C, Price EA, Sagoo MS, Onadim Z. Prognostic Information for Known Genetic Carriers of RB1 Pathogenic Variants (Germline and Mosaic). Ophthalmol Retina 2020; 5:381-387. [PMID: 32835838 DOI: 10.1016/j.oret.2020.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 08/17/2020] [Accepted: 08/18/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE To compare the number of tumors per eye for mosaic carriers of RB1 pathogenic variants with full germline variants and the conversion from unilateral to bilateral disease. DESIGN Retrospective cohort study comparing patients with retinoblastoma and different genetic subtypes: high penetrance (HP), low penetrance (LP), and mosaicism. PARTICIPANTS Data were analyzed between 1992 and 2018 at the Retinoblastoma Unit, Royal London Hospital, London, United Kingdom. All familial patients had a parent with a known pathogenic variant even if the parent did not manifest the disease. MAIN OUTCOME MEASURES Number of tumors per eye in children who developed retinoblastoma in that eye. Other outcomes included total number of tumors per patient, age at diagnosis, laterality at presentation and later, sex, and stage according to International Intraocular Retinoblastoma Classification. RESULTS A total of 111 patients were included: 64 full germline, familial patients (53 HP and 11 LP) and 47 mosaic patients. Twelve HP patients (23%) were unilateral, and 8 of 12 patients (67%) developed tumors in their previously unaffected eye. A total of 34 mosaic patients (72%) were unilateral, and only 2 (6%) developed tumors in their unaffected eye. Age at diagnosis was higher in mosaic patients (median, 22 months) than in HP patients (median 7) (P < 0.00002). The number of tumors per eye was fewer in patients with mosaic alleles (median, 1.0; range, 1-6) compared with patients with HP alleles (median, 3.0; range, 1-8) (P < 0.0003). All 3 children (4 eyes) with mosaicism and more than 2 tumors per eye had high levels of mosaicism. CONCLUSIONS Children with mosaic alleles have fewer tumors per eye compared with those with known high-penetrant pathogenic variants and are more likely to remain unilateral. The level of mosaicism has an impact on laterality and number of tumors.
Collapse
Affiliation(s)
- M Ashwin Reddy
- Retinoblastoma Service, Royal London Hospital, London, United Kingdom; NIHR Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital and University College London, Institute of Ophthalmology, London, United Kingdom.
| | - Mussa Butt
- Retinoblastoma Service, Royal London Hospital, London, United Kingdom
| | - Anne-Marie Hinds
- Retinoblastoma Service, Royal London Hospital, London, United Kingdom
| | - Catriona Duncan
- Retinoblastoma Service, Royal London Hospital, London, United Kingdom; Paediatric Oncology Department, Great Ormond Street Hospital, London, United Kingdom
| | - Elizabeth A Price
- Retinoblastoma Genetic Screening Unit, Royal London Hospital, London, United Kingdom
| | - Mandeep S Sagoo
- Retinoblastoma Service, Royal London Hospital, London, United Kingdom; NIHR Biomedical Research Centre for Ophthalmology at Moorfields Eye Hospital and University College London, Institute of Ophthalmology, London, United Kingdom
| | - Zerrin Onadim
- Retinoblastoma Genetic Screening Unit, Royal London Hospital, London, United Kingdom
| |
Collapse
|
18
|
Dittner-Moormann S, Reschke M, Biewald E, Kuechler A, Klein B, Timmermann B, Lohmann D, Ketteler P, Kanber D. 13q deletion syndrome resulting from balanced chromosomal rearrangement in father: the significance of parental karyotyping. Mol Cytogenet 2020; 13:31. [PMID: 32760450 PMCID: PMC7379829 DOI: 10.1186/s13039-020-00500-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/30/2020] [Indexed: 11/20/2022] Open
Abstract
Background Retinoblastoma is a malignancy of the eye in children characterized by biallelic inactivation of the retinoblastoma 1 gene (RB1), located at chromosome 13q14.2. Children with interstitial chromosome 13q deletions that include the RB1 gene show a predisposition to develop retinoblastoma and variable other features. Large 13q deletions with severe clinical phenotype are nearly always the result of a de novo mutation, i.e. the pathogenic alteration is not detected in parents. This results in a low risk for siblings to develop 13q deletion syndrome. Result Here, we describe a patient with profound muscle hypotonia, severe developmental delay and bilateral retinoblastoma carrying a large deletion in 13q13.3q14 with the size of 16 Mb, involving the RB1 gene. Neither parent showed retinoblastoma, muscle hypotonia or developmental delay. Chromosome analysis and Fluorescence in situ hybridization (FISH) showed a balanced complex chromosomal rearrangement (CCR) between chromosome 12 and 13 [ins(12;13)(q21.2;q12.3q14.3)] and an additional balanced translocation of chromosome 7 and 15 [t(7;15)(q31.2;q25.3)] in the healthy father. Malsegregation of the paternal insertional translocation involving chromosome 12 and 13 resulted in a 13q deletion syndrome of the child [46,XY,ins(12;13)(q21.2;q12.3q14.3)]. Conclusion Balanced translocations in parents are a rare cause of de novo RB1 deletions in offspring. This case report emphasizes the need for parental chromosomal analysis and FISH in parents of children diagnosed with 13q deletion syndrome or large RB1 gene deletions to precisely determine the recurrence risk in siblings. Guidelines for genetic testing should be revised accordingly.
Collapse
Affiliation(s)
- Sabine Dittner-Moormann
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Madlen Reschke
- Department of Pediatric Hematology, Oncology and Stem Cell Transplantation, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Eva Biewald
- Department of Ophthalmology, University Hospital Essen, Essen, Germany
| | - Alma Kuechler
- Institute of Human Genetics, University Hospital Essen, Essen, Germany
| | - Barbara Klein
- Sozialpädiatrisches Zentrum, Klinikum Frankfurt Höchst GmbH, Frankfurt a. M, Germany
| | - Beate Timmermann
- Department of Particle Therapy, West German Proton Therapy Centre Essen (WPE), West German Cancer Center (WTZ), German Cancer Consortium (DKTK), University Hospital Essen, Essen, Germany
| | - Dietmar Lohmann
- Institute of Human Genetics, University Hospital Essen, Essen, Germany
| | - Petra Ketteler
- Department of Pediatric Hematology and Oncology, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Deniz Kanber
- Institute of Human Genetics, University Hospital Essen, Essen, Germany
| |
Collapse
|
19
|
Lan X, Xu W, Tang X, Ye H, Song X, Lin L, Ren X, Yu G, Zhang H, Wu S. Spectrum of RB1 Germline Mutations and Clinical Features in Unrelated Chinese Patients With Retinoblastoma. Front Genet 2020; 11:142. [PMID: 32218800 PMCID: PMC7080181 DOI: 10.3389/fgene.2020.00142] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 02/06/2020] [Indexed: 11/13/2022] Open
Abstract
Retinoblastoma (Rb) is a primary intraocular malignant tumor that occurs primarily in children, and results from loss-of-function mutations in the RB transcriptional corepressor 1 (RB1) gene. Genetic testing forms the basis of genetic counseling for affected families, as well as for clinical management of this disease. The aim of this study was to identify germline RB1 mutations and correlate the identified mutations with the clinical features of Rb patients. Genomic DNA was isolated from peripheral blood of 180 unrelated Rb patients and their parents (118 unilaterally and 62 bilaterally affected probands). Mutations in the RB1 gene, including the promoter region and exons 1-27 with flanking intronic sequences, were identified by Sanger sequencing. The samples with negative sequencing results were further subjected to methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA) to detect gross deletions or duplications. Sixty-three distinct mutations were identified in 75 of the 180 (41.7%) probands. Of the 75 patients carrying RB1 mutations, 56 developed bilateral Rb, while 19 developed unilateral Rb. The total detection rates for bilateral and unilateral Rb were 90.3% (56/62) and 16.1% (19/118), respectively. Among the 75 patients, the spectrum of mutation types comprised 29.3% (22/75) nonsense mutations, 22.7% (17/75) splicing mutations, 17.3% (13/75) small insertions/deletions, 16.0% (12/75) large deletions/duplications, and 13.3% (10/75) missense mutations, while only 1% (1/75) of the mutations were in the promoter region of the RB1 gene. Age at diagnosis was significantly different (p < 0.01) between patients with positive and negative test results for germline RB1 mutations. A c.2359C > T mutation (p.R787X) was identified in identical twins, but one child was affected bilaterally and the other unilaterally. Of the five patients with deletion of the entire RB1 gene, the deletion of two patients was inherited from unaffected parents. In conclusion, in this study, we provide a comprehensive spectrum of RB1 germline mutations in Chinese Rb patients, and describe the correlations among RB1 mutations, age at diagnosis, and laterality; moreover, we report that the clinical features of individuals carrying an identical mutation in the RB1 gene were highly variable, indicating that the pathogenesis of Rb is more complicated than currently believed.
Collapse
Affiliation(s)
- Xiaoping Lan
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Wuhen Xu
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaojun Tang
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Haiyun Ye
- Department of Ophthalmology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaozhen Song
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Longlong Lin
- Department of Neurology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xiang Ren
- Department of Radiology, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Guangjun Yu
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hong Zhang
- Department of Clinical Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Shengnan Wu
- Molecular Diagnostic Laboratory, Shanghai Children's Hospital, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
20
|
Qadar LT, Baqari SAS, Maab H, Ali Asghar S, Hafeez MS. Recurrent Sporadic Bilateral Retinoblastoma in a Child with 13q Deletion Syndrome. Cureus 2020; 12:e6618. [PMID: 32064198 PMCID: PMC7008771 DOI: 10.7759/cureus.6618] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
13q syndrome is a chromosomal abnormality in which there is a pathognomic deletion of the genetic material on the long arm (q) of chromosome 13. Phenotypes of this syndrome are variable depending on the location of the deleted segment. The main manifestations of the syndrome include mental retardation, craniofacial dysmorphism, and increased susceptibility to tumors. We report a unique case of recurrent sporadic bilateral retinoblastoma (Rb) in a four-year-old boy carrying 13q (q12q14) interstitial deletion, which was treated successfully via enucleation and chemotherapy. Where most patients with familial Rb receive a single mutated Rb1 allele as the ‘first hit’, a small number of patients encounter interstitial deletion of the long arm of chromosome 13, resulting in the loss of the tumor suppressor Rb1 gene and presenting as sporadic cases.
Collapse
Affiliation(s)
- Laila Tul Qadar
- Internal Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Syed Ali Shazif Baqari
- Pediatric Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Center, Lahore, PAK
| | - Hira Maab
- Internal Medicine, Dow University of Health Sciences, Karachi, PAK
| | | | | |
Collapse
|
21
|
Tsutsumi M, Hattori H, Akita N, Maeda N, Kubota T, Horibe K, Fujita N, Kawai M, Shinkai Y, Kato M, Kato T, Kawamura R, Suzuki F, Kurahashi H. A female patient with retinoblastoma and severe intellectual disability carrying an X;13 balanced translocation without rearrangement in the RB1 gene: a case report. BMC Med Genomics 2019; 12:182. [PMID: 31806026 PMCID: PMC6896736 DOI: 10.1186/s12920-019-0640-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 11/29/2019] [Indexed: 12/02/2022] Open
Abstract
Background Female carriers of a balanced X; autosome translocation generally undergo selective inactivation of the normal X chromosome. This is because inactivation of critical genes within the autosomal region of the derivative translocation chromosome would compromise cellular function. We here report a female patient with bilateral retinoblastoma and a severe intellectual disability who carries a reciprocal X-autosomal translocation. Case presentation Cytogenetic and molecular analyses, a HUMARA (Human androgen receptor) assay, and methylation specific PCR (MSP) and bisulfite sequencing were performed using peripheral blood samples from the patient. The patient’s karyotype was 46,X,t(X;13)(q28;q14.1) by G-banding analysis. Further cytogenetic analysis located the entire RB1 gene and its regulatory region on der(X) with no translocation disruption. The X-inactivation pattern in the peripheral blood was highly skewed but not completely selected. MSP and deep sequencing of bisulfite-treated DNA revealed that an extensive 13q region, including the RB1 promoter, was unusually methylated in a subset of cells. Conclusions The der(X) region harboring the RB1 gene was inactivated in a subset of somatic cells, including the retinal cells, in the patient subject which acted as the first hit in the development of her retinoblastoma. In addition, the patient’s intellectual disability may be attributable to the inactivation of the der(X), leading to a 13q deletion syndrome-like phenotype, or to an active X-linked gene on der (13) leading to Xq28 functional disomy.
Collapse
Affiliation(s)
- Makiko Tsutsumi
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Hiroyoshi Hattori
- Department of Clinical Genetics, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan.,Department of Pediatrics, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Nobuhiro Akita
- Department of Pediatrics, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Naoko Maeda
- Department of Pediatrics, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Toshinobu Kubota
- Department of Ophthalmology, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Keizo Horibe
- Department of Pediatrics, National Hospital Organization, Nagoya Medical Center, Nagoya, Japan
| | - Naoko Fujita
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.,Genome and Transcriptome Analysis Center, Fujita Health University, Toyoake, Japan
| | - Miki Kawai
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Yasuko Shinkai
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Maki Kato
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Takema Kato
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Rie Kawamura
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan
| | - Fumihiko Suzuki
- Center for Collaboration in Research and Education, Fujita Health University, Toyoake, Japan
| | - Hiroki Kurahashi
- Division of Molecular Genetics, Institute for Comprehensive Medical Science, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi, 470-1192, Japan.
| |
Collapse
|
22
|
Nahon-Esteve S, Martel A, Maschi C, Caujolle JP, Baillif S, Lassalle S, Hofman P. The Molecular Pathology of Eye Tumors: A 2019 Update Main Interests for Routine Clinical Practice. Curr Mol Med 2019; 19:632-664. [DOI: 10.2174/1566524019666190726161044] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/12/2019] [Accepted: 07/15/2019] [Indexed: 12/17/2022]
Abstract
Over the last few years, we have seen constant development of molecular
pathology for the care of patients with cancer. The information obtained from molecular
data has transformed our thinking about the biological diversity of cancers, particularly in
the field of ophthalmic oncology. It has reoriented the way in which therapeutic decisions
and decisions concerning patient surveillance are made, both in the area of pediatric
cancers, including rhabdomyosarcoma and retinoblastoma, and adult cancers, such as
uveal melanoma and lymphomas. A better definition of the molecular classification of
these cancers and of the different biological pathways involved is essential to the
understanding of both the pathologist and the onco-ophthalmologist. Molecular tests
based on targeted or expanded analysis of gene panels are now available. These tests
can be performed with tumor tissue or biofluids (especially blood) to predict the
prognosis of tumors and, above all, the benefit of targeted therapies, immunotherapy or
even chemotherapy. Looking for the BAP1 mutation in uveal melanoma is essential
because of the associated metastatic risk. When treating retinoblastoma, it is mandatory
to assess the heritable status of RB1. Conjunctival melanoma requires investigation into
the BRAF mutation in the case of a locally advanced tumor. The understanding of
genomic alterations, the results of molecular tests and/or other biological tests predictive
of a therapeutic response, but also of the limits of these tests with respect to the
available biological resources, represents a major challenge for optimal patient
management in ophthalmic oncology. In this review, we present the current state of
knowledge concerning the different molecular alterations and therapeutic targets of
interest in ophthalmic oncology.
Collapse
Affiliation(s)
| | - Arnaud Martel
- Department of Ophthalmology, University Cote d'Azur, Nice, France
| | - Célia Maschi
- Department of Ophthalmology, University Cote d'Azur, Nice, France
| | | | | | - Sandra Lassalle
- Laboratory of Clinical and Experimental Pathology, University Cote d'Azur, Nice, France
| | - Paul Hofman
- Laboratory of Clinical and Experimental Pathology, University Cote d'Azur, Nice, France
| |
Collapse
|
23
|
Bornfeld N, Biewald E, Bauer S, Temming P, Lohmann D, Zeschnigk M. The Interdisciplinary Diagnosis and Treatment of Intraocular Tumors. DEUTSCHES ARZTEBLATT INTERNATIONAL 2019; 115:106-111. [PMID: 29510820 DOI: 10.3238/arztebl.2018.0106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 01/05/2017] [Accepted: 10/24/2017] [Indexed: 11/27/2022]
Abstract
BACKGROUND Recent years have seen major changes in the diagnosis and treatment of solid intraocular tumors, mainly owing to an improved molecular biological understanding of their pathogenesis, new therapeutic approaches for the local treatment of tumors in children, and long-term follow-up observations in clinical trials. METHODS This review is based on pertinent publications retrieved by a selective search in PubMed. RESULTS Retinoblastoma is the most common type of primary intraocular tumor, with approximately 8000 new cases per year around the world, while malignant melanoma of the uvea is the most common primary intraocular tumor in adults, with approximately 7000 new cases per year around the world. Intraocular metastases of malignant tumors are ten times more common, in terms of incidence, than primary intraocular tumors and are therefore the most common intraocular tumors overall. Improved methods of intraocular biopsy, diagnostic imaging, and molecular genetic investigation have led to steady improvement in clinical and predictive diagnostic assessment. In the treatment of retinoblastoma, local techniques including brachytherapy and intra-arterial and intravitreal chemotherapy play a prominent role. Prognostic molecular-genetic testing now enables the highly selective identification of uveal melanomas that have a high potential to metastasize. Cutaneous and uveal melanomas differ both in their clinical behavior and in their basic biological features; to date, effective systemic treatment has been established for melanoma of the skin, but not for metastatic melanoma of the uvea. Intraocular metastases are common and often the initial manifestation of an extraocular tumor, particularly lung cancer. CONCLUSION Modern diagnostic and therapeutic concepts for intraocular tumors can only be implemented through the close interdisciplinary collaboration of ophthal - mologists, oncologists, radiologists, radiotherapists, pathologists, and human geneticists.
Collapse
Affiliation(s)
- Norbert Bornfeld
- Department of Ophthalmology, Tumor Center Western Germany, Essen University Hospital; Department of Internal Medicine (Tumor Research), Tumor Center Western Germany, Essen University Hospital; Department of Pediatrics III, Tumor Center Western Germany, Essen University Hospital; Institute for Human Genetics, Tumor Center Western Germany, Essen University Hospital
| | | | | | | | | | | |
Collapse
|
24
|
Munier FL, Beck-Popovic M, Chantada GL, Cobrinik D, Kivelä TT, Lohmann D, Maeder P, Moll AC, Carcaboso AM, Moulin A, Schaiquevich P, Bergin C, Dyson PJ, Houghton S, Puccinelli F, Vial Y, Gaillard MC, Stathopoulos C. Conservative management of retinoblastoma: Challenging orthodoxy without compromising the state of metastatic grace. "Alive, with good vision and no comorbidity". Prog Retin Eye Res 2019; 73:100764. [PMID: 31173880 DOI: 10.1016/j.preteyeres.2019.05.005] [Citation(s) in RCA: 123] [Impact Index Per Article: 24.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2019] [Revised: 05/25/2019] [Accepted: 05/29/2019] [Indexed: 12/21/2022]
Abstract
Retinoblastoma is lethal by metastasis if left untreated, so the primary goal of therapy is to preserve life, with ocular survival, visual preservation and quality of life as secondary aims. Historically, enucleation was the first successful therapeutic approach to decrease mortality, followed over 100 years ago by the first eye salvage attempts with radiotherapy. This led to the empiric delineation of a window for conservative management subject to a "state of metastatic grace" never to be violated. Over the last two decades, conservative management of retinoblastoma witnessed an impressive acceleration of improvements, culminating in two major paradigm shifts in therapeutic strategy. Firstly, the introduction of systemic chemotherapy and focal treatments in the late 1990s enabled radiotherapy to be progressively abandoned. Around 10 years later, the advent of chemotherapy in situ, with the capitalization of new routes of targeted drug delivery, namely intra-arterial, intravitreal and now intracameral injections, allowed significant increase in eye preservation rate, definitive eradication of radiotherapy and reduction of systemic chemotherapy. Here we intend to review the relevant knowledge susceptible to improve the conservative management of retinoblastoma in compliance with the "state of metastatic grace", with particular attention to (i) reviewing how new imaging modalities impact the frontiers of conservative management, (ii) dissecting retinoblastoma genesis, growth patterns, and intraocular routes of tumor propagation, (iii) assessing major therapeutic changes and trends, (iv) proposing a classification of relapsing retinoblastoma, (v) examining treatable/preventable disease-related or treatment-induced complications, and (vi) appraising new therapeutic targets and concepts, as well as liquid biopsy potentiality.
Collapse
Affiliation(s)
- Francis L Munier
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland.
| | - Maja Beck-Popovic
- Unit of Pediatric Hematology-Oncology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Guillermo L Chantada
- Hemato-Oncology Service, Hospital JP Garrahan, Buenos Aires, Argentina; Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain; Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - David Cobrinik
- The Vision Center and The Saban Research Institute, Children's Hospital Los Angeles, Los Angeles, CA, USA; USC Roski Eye Institute, Department of Biochemistry & Molecular Medicine, Norris Comprehensive Cancer Center, Keck School of Medicine of the University of Southern California, Los Angeles, CA, USA
| | - Tero T Kivelä
- Department of Ophthalmology, Ocular Oncology and Pediatric Ophthalmology Services, Helsinki University Hospital, Helsinki, Finland
| | - Dietmar Lohmann
- Eye Oncogenetics Research Group, Institute of Human Genetics, University Hospital Essen, Essen, Germany
| | - Philippe Maeder
- Unit of Neuroradiology, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Annette C Moll
- UMC, Vrije Universiteit Amsterdam, Department of Ophthalmology, Cancer Center Amsterdam, Amsterdam, Netherlands
| | - Angel Montero Carcaboso
- Pediatric Hematology and Oncology, Hospital Sant Joan de Deu, Barcelona, Spain; Institut de Recerca Sant Joan de Deu, Barcelona, Spain
| | - Alexandre Moulin
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Paula Schaiquevich
- Unit of Clinical Pharmacokinetics, Hospital de Pediatria JP Garrahan, Buenos Aires, Argentina; National Scientific and Technical Research Council (CONICET), Buenos Aires, Argentina
| | - Ciara Bergin
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Paul J Dyson
- Institut des Sciences et Ingénierie Chimiques, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015, Lausanne, Switzerland
| | - Susan Houghton
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Francesco Puccinelli
- Interventional Neuroradiology Unit, Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland
| | - Yvan Vial
- Materno-Fetal Medicine Unit, Woman-Mother-Child Department, University Hospital of Lausanne, Switzerland
| | - Marie-Claire Gaillard
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| | - Christina Stathopoulos
- Jules-Gonin Eye Hospital, Fondation Asile des Aveugles, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
25
|
Bellucco FT, Rodrigues de Oliveira-Júnior H, Santos Guilherme R, Bragagnolo S, Alvarez Perez AB, Ayres Meloni V, Melaragno MI. Deletion of Chromosome 13 due to Different Rearrangements and Impact on Phenotype. Mol Syndromol 2019; 10:139-146. [PMID: 31191202 DOI: 10.1159/000497402] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2019] [Indexed: 12/20/2022] Open
Abstract
Patients with deletion of chromosome 13 present with variable clinical features, and the correlation between phenotype and genomic aberration is not well established in the literature, mainly due to variable sizes of the deleted segments and inaccuracy of breakpoint mapping. In order to improve the genotype-phenotype correlation, we obtained clinical and cytogenomic data from 5 Brazilian patients with different chromosome 13 deletions characterized by G-banding and array techniques. Breakpoints were nonrecurrent, with deletion sizes ranging from 3.8 to 43.3 Mb. Our patients showed some classic features associated with 13q deletion, independent of the location and size of the deletion: hypotonia, growth delay, psychomotor developmental delay, microcephaly, central nervous system anomalies, and minor facial dysmorphism as well as urogenital and limb abnormalities. Comparisons between the literature and our patients' data allowed us to narrow the critical regions that were previously reported for microphthalmia and urogenital abnormalities, indicating that gene haploinsufficiency of ARHGEF7, PCDH9 and DIAPH3, of MIR17HG and GPC6, and of EFNB2 may contribute to microcephaly, cardiovascular disease, and urogenital abnormalities, respectively. The knowledge about genes involved in the phenotypic features found in 13q deletion patients may help us to understand how the genes interact and contribute to their clinical phenotype, improving the patient's clinical follow-up.
Collapse
Affiliation(s)
- Fernanda T Bellucco
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | | | - Roberta Santos Guilherme
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Silvia Bragagnolo
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana B Alvarez Perez
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Vera Ayres Meloni
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Maria I Melaragno
- Genetics Division, Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
George A, Venkatesan S, Ashok N, Saraswathy R, Hande MP. Assessment of genomic instability and proliferation index in cultured lymphocytes of patients with Down syndrome, congenital anomalies and aplastic anaemia. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 836:98-103. [DOI: 10.1016/j.mrgentox.2018.06.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/11/2018] [Revised: 05/17/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023]
|
27
|
A novel de novo heterozygous deletion at 13q14.2-q21.1 in two siblings with mild intellectual disability. GENE REPORTS 2018. [DOI: 10.1016/j.genrep.2018.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
28
|
Shah S, Koban Y, Le BHA, Bechtold M, Zolfaghari E, Kim JW, Berry JL. Iris Hypoplasia as the Presenting Sign of Retinoblastoma in a Child With a 13q Deletion. J Pediatr Ophthalmol Strabismus 2018; 55:e10-e13. [PMID: 29684226 PMCID: PMC7444716 DOI: 10.3928/01913913-20180215-02] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 02/02/2018] [Indexed: 11/20/2022]
Abstract
Germline partial chromosomal deletions of the entire RB1 gene (13q deletions), account for 6% of the RB1 mutational spectrum. The authors report the rare case of one patient with suspected bilateral iris heterochromia who actually had iris hypoplasia (often masquerading as heterochromia) and bilateral retinoblastoma, due to 13q deletion syndrome. [J Pediatr Ophthalmol. 2018;55:e10-e13.].
Collapse
|
29
|
Asnaghi L, Tripathy A, Yang Q, Kaur H, Hanaford A, Yu W, Eberhart CG. Targeting Notch signaling as a novel therapy for retinoblastoma. Oncotarget 2018; 7:70028-70044. [PMID: 27661116 PMCID: PMC5342532 DOI: 10.18632/oncotarget.12142] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Accepted: 09/14/2016] [Indexed: 01/15/2023] Open
Abstract
Retinoblastoma is the most common intraocular malignancy of childhood. Notch plays a key role in retinal cells from which retinoblastomas arise, and we therefore studied the role of Notch signaling in promoting retinoblastoma proliferation. Moderate or strong nuclear expression of Hes1 was found in 10 of 11 human retinoblastoma samples analyzed immunohistochemically, supporting a role for Notch in retinoblastoma growth. Notch pathway components were present in WERI Rb1 and Y79 retinoblastoma lines, with Jag2 and DLL4 more highly expressed than other ligands, and Notch1 and Notch2 more abundant than Notch3. The cleaved/active form of Notch1 was detectable in both lines. Inhibition of the pathway, achieved using a γ-secretase inhibitor (GSI) or by downregulating Jag2, DLL4 or CBF1 using short hairpin RNA, potently reduced growth, proliferation and clonogenicity in both lines. Upregulation of CXCR4 and CXCR7 and downregulation of PI3KC2β were identified by microarray upon Jag2 suppression. The functional importance of PI3KC2β was confirmed using shRNA. Synergy was found by combining GSI with Melphalan at their IC50. These findings indicate that Notch pathway is active in WERI Rb1 and Y79, and in most human retinoblastoma samples, and suggest that Notch antagonists may represent a new approach to more effectively treat retinoblastoma.
Collapse
Affiliation(s)
- Laura Asnaghi
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Arushi Tripathy
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Qian Yang
- Department of Ophthalmology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Harpreet Kaur
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Allison Hanaford
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Wayne Yu
- Microarray Core Facility, Sidney Kimmel Cancer Center, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| | - Charles G Eberhart
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.,Department of Ophthalmology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA.,Department of Oncology, Johns Hopkins University, School of Medicine, Baltimore, MD, USA
| |
Collapse
|
30
|
Lowther C, Merico D, Costain G, Waserman J, Boyd K, Noor A, Speevak M, Stavropoulos DJ, Wei J, Lionel AC, Marshall CR, Scherer SW, Bassett AS. Impact of IQ on the diagnostic yield of chromosomal microarray in a community sample of adults with schizophrenia. Genome Med 2017; 9:105. [PMID: 29187259 PMCID: PMC5708103 DOI: 10.1186/s13073-017-0488-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 11/01/2017] [Indexed: 11/10/2022] Open
Abstract
Background Schizophrenia is a severe psychiatric disorder associated with IQ deficits. Rare copy number variations (CNVs) have been established to play an important role in the etiology of schizophrenia. Several of the large rare CNVs associated with schizophrenia have been shown to negatively affect IQ in population-based controls where no major neuropsychiatric disorder is reported. The aim of this study was to examine the diagnostic yield of microarray testing and the functional impact of genome-wide rare CNVs in a community ascertained cohort of adults with schizophrenia and low (< 85) or average (≥ 85) IQ. Methods We recruited 546 adults of European ancestry with schizophrenia from six community psychiatric clinics in Canada. Each individual was assigned to the low or average IQ group based on standardized tests and/or educational attainment. We used rigorous methods to detect genome-wide rare CNVs from high-resolution microarray data. We compared the burden of rare CNVs classified as pathogenic or as a variant of unknown significance (VUS) between each of the IQ groups and the genome-wide burden and functional impact of rare CNVs after excluding individuals with a pathogenic CNV. Results There were 39/546 (7.1%; 95% confidence interval [CI] = 5.2–9.7%) schizophrenia participants with at least one pathogenic CNV detected, significantly more of whom were from the low IQ group (odds ratio [OR] = 5.01 [2.28–11.03], p = 0.0001). Secondary analyses revealed that individuals with schizophrenia and average IQ had the lowest yield of pathogenic CNVs (n = 9/325; 2.8%), followed by those with borderline intellectual functioning (n = 9/130; 6.9%), non-verbal learning disability (n = 6/29; 20.7%), and co-morbid intellectual disability (n = 15/62; 24.2%). There was no significant difference in the burden of rare CNVs classified as a VUS between any of the IQ subgroups. There was a significantly (p=0.002) increased burden of rare genic duplications in individuals with schizophrenia and low IQ that persisted after excluding individuals with a pathogenic CNV. Conclusions Using high-resolution microarrays we were able to demonstrate for the first time that the burden of pathogenic CNVs in schizophrenia differs significantly between IQ subgroups. The results of this study have implications for clinical practice and may help inform future rare variant studies of schizophrenia using next-generation sequencing technologies. Electronic supplementary material The online version of this article (doi:10.1186/s13073-017-0488-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Chelsea Lowther
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1.,Institute of Medical Science, University of Toronto, Toronto, ON, Canada
| | - Daniele Merico
- Deep Genomics Inc, Toronto, ON, Canada.,The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Gregory Costain
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1.,Division of Clinical and Metabolic Genetics, The Hospital for Sick Children, Toronto, ON, Canada
| | | | - Kerry Boyd
- Department of Psychiatry & Behavioural Neurosciences, McMaster University, Hamilton, ON, Canada
| | - Abdul Noor
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Marsha Speevak
- Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | | | - John Wei
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Anath C Lionel
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Christian R Marshall
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada.,Genome Diagnostics, Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
| | - Stephen W Scherer
- The Centre for Applied Genomics and Program in Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.,McLaughlin Centre, University of Toronto, Toronto, ON, Canada
| | - Anne S Bassett
- Clinical Genetics Research Program, Centre for Addiction and Mental Health, 33 Russell Street, Room 1100, Toronto, ON, Canada, M5S 2S1. .,Institute of Medical Science, University of Toronto, Toronto, ON, Canada. .,Toronto General Research Institute, University Health Network, Toronto, ON, Canada. .,Cambell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, ON, Canada. .,Department of Psychiatry, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
31
|
Avior Y, Lezmi E, Yanuka D, Benvenisty N. Modeling Developmental and Tumorigenic Aspects of Trilateral Retinoblastoma via Human Embryonic Stem Cells. Stem Cell Reports 2017; 8:1354-1365. [PMID: 28392220 PMCID: PMC5425613 DOI: 10.1016/j.stemcr.2017.03.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Revised: 03/06/2017] [Accepted: 03/07/2017] [Indexed: 12/18/2022] Open
Abstract
Human embryonic stem cells (hESCs) provide a platform for studying human development and understanding mechanisms underlying diseases. Retinoblastoma-1 (RB1) is a key regulator of cell cycling, of which biallelic inactivation initiates retinoblastoma, the most common congenital intraocular malignancy. We developed a model to study the role of RB1 in early development and tumor formation by generating RB1-null hESCs using CRISPR/Cas9. RB1−/− hESCs initiated extremely large teratomas, with neural expansions similar to those of trilateral retinoblastoma tumors, in which retinoblastoma is accompanied by intracranial neural tumors. Teratoma analysis further revealed a role for the transcription factor ZEB1 in RB1-mediated ectoderm differentiation. Furthermore, RB1−/− cells displayed mitochondrial dysfunction similar to poorly differentiated retinoblastomas. Screening more than 100 chemotherapies revealed an RB1–/–-specific cell sensitivity to carboplatin, exploiting their mitochondrial dysfunction. Together, our work provides a human pluripotent cell model for retinoblastoma and sheds light on developmental and tumorigenic roles of RB1. RB1-null hESCs were generated using CRISPR/Cas9 RB1−/− hESCs generate large, neural-enriched teratomas, possibly by ZEB1 activation RB1 inactivation triggers aberrant mitochondrial abundance and function Unbiased drug screening found that carboplatin specifically targets RB1-null cells
Collapse
Affiliation(s)
- Yishai Avior
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University of Jerusalem, Givat-Ram, Jerusalem 91904, Israel
| | - Elyad Lezmi
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University of Jerusalem, Givat-Ram, Jerusalem 91904, Israel
| | - Dorit Yanuka
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University of Jerusalem, Givat-Ram, Jerusalem 91904, Israel
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Institute of Life Sciences, The Hebrew University of Jerusalem, Givat-Ram, Jerusalem 91904, Israel.
| |
Collapse
|
32
|
Scollon S, Anglin AK, Thomas M, Turner JT, Wolfe Schneider K. A Comprehensive Review of Pediatric Tumors and Associated Cancer Predisposition Syndromes. J Genet Couns 2017; 26:387-434. [PMID: 28357779 DOI: 10.1007/s10897-017-0077-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2016] [Accepted: 01/30/2017] [Indexed: 12/11/2022]
Abstract
An understanding of the role of inherited cancer predisposition syndromes in pediatric tumor diagnoses continues to develop as more information is learned through the application of genomic technology. Identifying patients and their relatives at an increased risk for developing cancer is an important step in the care of this patient population. The purpose of this review is to highlight various tumor types that arise in the pediatric population and the cancer predisposition syndromes associated with those tumors. The review serves as a guide for recognizing genes and conditions to consider when a pediatric cancer referral presents to the genetics clinic.
Collapse
Affiliation(s)
- Sarah Scollon
- Department of Pediatrics, Baylor College of Medicine, Texas Children's Cancer Center, Texas Children's Hospital, 1102 Bates St, FC 1200, Houston, TX, 77030, USA.
| | | | | | - Joyce T Turner
- Department of Genetics and Metabolism, Children's National Medical Center, Washington, DC, USA
| | - Kami Wolfe Schneider
- Department of Pediatrics, University of Colorado, Center for Cancer and Blood Disorders, Children's Hospital Colorado, Aurora, CO, USA
| |
Collapse
|
33
|
Soliman SE, Racher H, Zhang C, MacDonald H, Gallie BL. Genetics and Molecular Diagnostics in Retinoblastoma--An Update. Asia Pac J Ophthalmol (Phila) 2017; 6:197-207. [PMID: 28399338 DOI: 10.22608/apo.201711] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Accepted: 03/09/2017] [Indexed: 11/08/2022] Open
Abstract
Retinoblastoma is the prototype genetic cancer: in one or both eyes of young children, most retinoblastomas are initiated by biallelic mutation of the retinoblastoma tumor suppressor gene, RB1, in a developing retinal cell. All those with bilateral retinoblastoma have heritable cancer, although 95% have not inherited the RB1 mutation. Non-heritable retinoblastoma is always unilateral, with 98% caused by loss of both RB1 alleles from the tumor, whereas 2% have normal RB1 in tumors initiated by amplification of the MYCN oncogene. Good understanding of retinoblastoma genetics supports optimal care for retinoblastoma children and their families. Retinoblastoma is the first cancer to officially acknowledge the seminal role of genetics in cancer, by incorporating "H" into the eighth edition of cancer staging (2017): those who carry the RB1 cancer-predisposing gene are H1; those proven to not carry the familial RB1 mutation are H0; and those at unknown risk are HX. We suggest H0* be used for those with residual <1% risk to carry a RB1 mutation due to undetectable mosaicism. Loss of RB1 from a susceptible developing retinal cell initiates the benign precursor, retinoma. Progressive genomic changes result in retinoblastoma, and cancer progression ensues with increasing genomic disarray. Looking forward, novel therapies are anticipated from studies of retinoblastoma and metastatic tumor cells and the second primary cancers that the carriers of RB1 mutations are at high risk to develop. Here, we summarize the concepts of retinoblastoma genetics for ophthalmologists in a question/answer format to assist in the care of patients and their families.
Collapse
Affiliation(s)
- Sameh E Soliman
- Department of Ophthalmology and Vision Sciences, University of Toronto, Ontario, Canada
- Department of Ophthalmology, Faculty of Medicine, University of Alexandria, Alexandria, Egypt
| | | | - Chengyue Zhang
- Department of Ophthalmology, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Heather MacDonald
- Department of Ophthalmology and Vision Sciences, University of Toronto, Ontario, Canada
| | - Brenda L Gallie
- Department of Ophthalmology and Vision Sciences, University of Toronto, Ontario, Canada
- Departments of Ophthalmology, Molecular Genetics, and Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
34
|
Matsui T, Nieto-Estévez V, Kyrychenko S, Schneider JW, Hsieh J. Retinoblastoma protein controls growth, survival and neuronal migration in human cerebral organoids. Development 2017; 144:1025-1034. [PMID: 28087635 DOI: 10.1242/dev.143636] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2016] [Accepted: 12/16/2016] [Indexed: 01/22/2023]
Abstract
The tumor suppressor retinoblastoma protein (RB) regulates S-phase cell cycle entry via E2F transcription factors. Knockout (KO) mice have shown that RB plays roles in cell migration, differentiation and apoptosis, in developing and adult brain. In addition, the RB family is required for self-renewal and survival of human embryonic stem cells (hESCs). Since little is known about the role of RB in human brain development, we investigated its function in cerebral organoids differentiated from gene-edited hESCs lacking RB. We show that RB is abundantly expressed in neural stem and progenitor cells in organoids at 15 and 28 days of culture. RB loss promoted S-phase entry in DCX+ cells and increased apoptosis in Sox2+ neural stem and progenitor cells, and in DCX+ and Tuj1+ neurons. Associated with these cell cycle and pro-apoptotic effects, we observed increased CCNA2 and BAX gene expression, respectively. Moreover, we observed aberrant Tuj1+ neuronal migration in RB-KO organoids and upregulation of the gene encoding VLDLR, a receptor important in reelin signaling. Corroborating the results in RB-KO organoids in vitro, we observed ectopically localized Tuj1+ cells in RB-KO teratomas grown in vivo Taken together, these results identify crucial functions for RB in the cerebral organoid model of human brain development.
Collapse
Affiliation(s)
- Takeshi Matsui
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Vanesa Nieto-Estévez
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Sergii Kyrychenko
- Department of Internal Medicine and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jay W Schneider
- Department of Internal Medicine and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jenny Hsieh
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
35
|
Truong DT, Shriberg LD, Smith SD, Chapman KL, Scheer-Cohen AR, DeMille MMC, Adams AK, Nato AQ, Wijsman EM, Eicher JD, Gruen JR. Multipoint genome-wide linkage scan for nonword repetition in a multigenerational family further supports chromosome 13q as a locus for verbal trait disorders. Hum Genet 2016; 135:1329-1341. [PMID: 27535846 PMCID: PMC5065602 DOI: 10.1007/s00439-016-1717-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 07/22/2016] [Indexed: 12/19/2022]
Abstract
Verbal trait disorders encompass a wide range of conditions and are marked by deficits in five domains that impair a person's ability to communicate: speech, language, reading, spelling, and writing. Nonword repetition is a robust endophenotype for verbal trait disorders that is sensitive to cognitive processes critical to verbal development, including auditory processing, phonological working memory, and motor planning and programming. In the present study, we present a six-generation extended pedigree with a history of verbal trait disorders. Using genome-wide multipoint variance component linkage analysis of nonword repetition, we identified a region spanning chromosome 13q14-q21 with LOD = 4.45 between 52 and 55 cM, spanning approximately 5.5 Mb on chromosome 13. This region overlaps with SLI3, a locus implicated in reading disability in families with a history of specific language impairment. Our study of a large multigenerational family with verbal trait disorders further implicates the SLI3 region in verbal trait disorders. Future studies will further refine the specific causal genetic factors in this locus on chromosome 13q that contribute to language traits.
Collapse
Affiliation(s)
- D T Truong
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - L D Shriberg
- Waisman Center, University of Wisconsin-Madison, Madison, WI, 53705, USA
| | - S D Smith
- Department of Pediatrics, University of Nebraska at Omaha, Omaha, NE, 68182, USA
| | - K L Chapman
- Department of Communication Sciences and Disorders, University of Utah, Salt Lake City, UT, 84112, USA
| | - A R Scheer-Cohen
- Department of Speech-Language Pathology, California State University, San Marcos, CA, 92096, USA
| | - M M C DeMille
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - A K Adams
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - A Q Nato
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, 98195, USA
| | - E M Wijsman
- Division of Medical Genetics, Department of Medicine, University of Washington, Seattle, WA, 98195, USA
- Department of Biostatistics and Department of Genome Sciences, University of Washington, Seattle, WA, 98195, USA
| | - J D Eicher
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA
| | - J R Gruen
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, 06510, USA.
- Department of Genetics, Yale School of Medicine, New Haven, CT, 06510, USA.
- Investigative Medicine Program, Yale School of Medicine, New Haven, CT, 06510, USA.
| |
Collapse
|
36
|
Brennan RC, Qaddoumi I, Billups CA, Kaluzny T, Furman WL, Wilson MW. Patients with retinoblastoma and chromosome 13q deletions have increased chemotherapy-related toxicities. Pediatr Blood Cancer 2016; 63:1954-8. [PMID: 27409525 PMCID: PMC5683082 DOI: 10.1002/pbc.26138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Revised: 06/13/2016] [Accepted: 06/14/2016] [Indexed: 01/01/2023]
Abstract
BACKGROUND A total of 5-10% of patients with retinoblastoma (RB) harbor deletion of the long arm (q) chromosome 13 (13q-). The treatment-related toxicities in this population have not been described. METHODS Sixty-eight RB patients on a single institutional protocol (RET5) from 2005 to 2010 were reviewed. Genetic screening identified 11 patients (seven female) with 13q-. Patients with early (Reese-Ellsworth [R-E] group I-III) disease (6/23 with 13q-) received eight courses of vincristine/carboplatin (VC). Patients with advanced (R-E group IV-V) bilateral disease (2/27 with 13q-) received two courses of vincristine/topotecan (VT) followed by nine courses of alternating VT/VC. Patients undergoing upfront enucleation received histopathology-based chemotherapy: intermediate risk (2/8 with 13q-) or high risk (1/10 with 13q-). Dose reductions were mandated for >7 day delay in two consecutive courses following hematologic toxicity. Grades 3 and 4 hematologic, infectious, and gastrointestinal toxicities were compared between RET5 patients with and without 13q-. RESULTS Demographics were similar between groups. When present, prolonged neutropenia (median 7 days, range 0-14 days) delayed chemotherapy and resulted in more frequent dose reductions among 13q- patients (5/11) than non-13q- patients (4/57) (P < 0.01). GI toxicity was similar between groups (5/11 13q- vs. 13/57 non-13q-; P = 0.14), but halted chemotherapy in one 13q- patient. Infectious complications and disease outcomes were similar between groups. At follow-up, all patients are alive (median 6.1 years, range 7.6 months-9.5 years). CONCLUSIONS 13q- RB patients had a higher incidence of neutropenia requiring chemotherapy dose reductions, but did not have increased treatment failure.
Collapse
Affiliation(s)
- RC Brennan
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA,Department of Pediatrics, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA,Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| | - I Qaddoumi
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA,Department of Pediatrics, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| | - CA Billups
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - T Kaluzny
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - WL Furman
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA,Department of Pediatrics, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| | - MW Wilson
- Department of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA,Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, USA,Department of Ophthalmology, University of Tennessee Health Science Center, College of Medicine, Memphis, TN, USA
| |
Collapse
|
37
|
A del(13)(q21.32q31.2)dn refined to 21.9 Mb in a female toddler with irides heterochromia and hypopigmentation: appraisal of interstitial mid-13q deletions. Clin Dysmorphol 2016; 26:33-37. [PMID: 27750267 DOI: 10.1097/mcd.0000000000000159] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
38
|
Akopyan HR, Kushniruk VO, Mykytenko DO, Huleuk NL, Kremenskaya Y, Lukash LL. Chromosomal DNA balance in human stem cell line 4BL. CYTOL GENET+ 2016. [DOI: 10.3103/s0095452716040022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
39
|
Seppänen VI, Artama MS, Malila NK, Pitkäniemi JM, Rantanen ME, Ritvanen AK, Madanat-Harjuoja LM. Risk for congenital anomalies in offspring of childhood, adolescent and young adult cancer survivors. Int J Cancer 2016; 139:1721-30. [PMID: 27280956 DOI: 10.1002/ijc.30226] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 05/11/2016] [Accepted: 05/27/2016] [Indexed: 11/06/2022]
Abstract
Offspring of cancer survivors (CS) may be at risk for congenital anomalies due to the mutagenic therapies received by their parents. Our population-based cohort study aimed to investigate the risk for congenital anomalies in offspring of CS compared to offspring of their siblings. Using the Finnish Cancer Registry, Central Population Register, and Hospital Discharge Register, we identified hospital contacts due to congenital anomalies in 6,862 offspring of CS (early-onset cancer between 1953 and 2004) and 35,690 offspring of siblings. Associations between congenital anomalies and cancer were evaluated using generalized linear regression modelling. The ratio of congenital anomalies in offspring of CS (3.2%) was slightly, but non-significantly, elevated compared to that in offspring of siblings (2.7%) [prevalence ratio (PR) 1.07, 95% confidence interval (CI) 0.91-1.25]. When offspring of childhood and adolescent survivors (0-19 years at cancer diagnosis) were compared to siblings' offspring, the risk for congenital anomalies was non-significantly increased (PR 1.17, 95% CI 0.92-1.49). No such increase existed for offspring of young adult survivors (20-34 years at cancer diagnosis) (PR 1.01, 95% CI 0.83-1.23). The risks for congenital anomalies were elevated among offspring of CS diagnosed with cancer in the earlier decades (1955-1964: PR 2.77, 95% C I 1.26-6.11; and 1965-1974: PR 1.55, 95% C I 0.94-2.56). In our study, we did not detect an overall elevated risk for congenital anomalies in offspring of survivors diagnosed in young adulthood. An association between cancer exposure of the parent and congenital anomalies in the offspring appeared only for those CS who were diagnosed in the earlier decades.
Collapse
Affiliation(s)
- Viivi I Seppänen
- University of Helsinki, Helsinki, Finland.,Finnish Cancer Registry, Helsinki, Finland
| | | | - Nea K Malila
- Finnish Cancer Registry, Helsinki, Finland.,School of Health Sciences, University of Tampere, Tampere, Finland
| | | | | | - Annukka K Ritvanen
- Finnish Register of Congenital Malformations, THL National Institute for Health and Welfare, Helsinki, Finland
| | - Laura-Maria Madanat-Harjuoja
- Finnish Cancer Registry, Helsinki, Finland.,Department of Pediatrics, Jorvi Hospital and University of Helsinki, Helsinki, Finland
| |
Collapse
|
40
|
Sagi M, Frenkel A, Eilat A, Weinberg N, Frenkel S, Pe'er J, Abeliovich D, Lerer I. Genetic screening in patients with Retinoblastoma in Israel. Fam Cancer 2016; 14:471-80. [PMID: 25754945 DOI: 10.1007/s10689-015-9794-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Retinoblastoma (Rb) is a childhood tumor (~1 in 20,000 live births) developing in the retina due to mutations in the RB1 gene. Identification of the oncogenic mutations in the RB1 gene is important for the clinical management and for genetic counseling to families with a child or a parent affected with the tumor. Here we present our experience in detecting the pathogenic mutations in blood samples, from 150 unrelated Rb patients and highlight the relevant counseling issues. Mutation screening in the RB1 gene was based on Sanger sequencing, mosaicism of recurrent CpG transition mutations was detected by allele specific PCR and multiplex ligation dependent probe amplification for detecting of large deletions/duplications. The overall detection rate of mutations in our cohort was 55% (82/150). In the familial cases it was 100% (17/17), in bilateral and unilateral-multifocal sporadic cases 91% (50/55), and in the unilateral sporadic cases 19% (15/78). Nonsense mutations and small deletions or insertions that results in transcripts with premature termination codons that are subject to nonsense mediated decay were the most frequent, detected in 50/82 (61%) of the patients. The rest were large deletions detected in 14/82 (17%), splice site mutations detected in 11/82 (13%), missense mutations in four patients and mutations in the promoter sequence in three patients. Mutation mosaicism ranging from 10 to 30% was detected by allele specific PCR in ten patients, 9% (5/55) of patients with bilateral tumor and 33% (5/15) of the patients with unilateral tumor. In three patients rare variants were detected as the only finding which was also detected in other healthy family members. Allele specific amplification of recurrent mutations raises in our cohort the identification rate from 82 to 91% in the sporadic bilateral cases and from 13 to 19% in the unilateral sporadic cases. Most mosaic cases could not be identified by Sanger sequencing and therefore screening for recurrent CpG transition mutations by allele specific amplification is of utmost importance. Molecular screening is important for the genetic counseling regarding the risk for tumor development and the relevance for prenatal diagnosis but in several families is accompanied by detecting rare variants that might be rare polymorphisms or low penetrant mutations.
Collapse
Affiliation(s)
- Michal Sagi
- Department of Human Genetics and Metabolic Diseases, Hadassah-Hebrew University Medical Center, Kiryat Hadassah, POB 12000, 91120, Jerusalem, Israel
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Sparrow J, Brennan R, Mao S, Ness KK, Rodriguez-Galindo C, Wilson M, Qaddoumi I. Participation in an occupational therapy referral program for children with retinoblastoma. J Pediatr Rehabil Med 2016; 9:117-24. [PMID: 27285804 PMCID: PMC4939593 DOI: 10.3233/prm-160372] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
PURPOSE Because retinoblastoma typically arises at a very young age, children are particularly vulnerable to vision impairment, associated developmental delays, and functional limitations. Limited information is available describing developmental delay and functional limitations in this population, necessitating supportive services including rehabilitation. The aims of this study were to describe the participation of children with newly diagnosed retinoblastoma in an occupational therapy program that identifies children in need of rehabilitation services. We also identify indications for referral to rehabilitation services among children with newly diagnosed retinoblastoma and enumerate the likelihood of these children receiving the recommended services. METHODS Twenty-two children participated in longitudinal occupational therapy assessments during the first year after diagnosis. RESULTS We recommended 1 or more types of rehabilitation services for 16 of 22 (72.7%) participants. Twelve of 16 (75%) received services. CONCLUSIONS The results of this pilot study indicate that implementing a prospective occupational therapy-screening program is feasible and results in identification and initiation of therapy services in some children with retinoblastoma. Developmental screenings and follow-up of children with retinoblastoma is strongly recommended.
Collapse
Affiliation(s)
- Jessica Sparrow
- Rehabilitation Services, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rachel Brennan
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Shenghua Mao
- Department of Biostatistics, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Kirsten K Ness
- Department of Epidemiology & Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Carlos Rodriguez-Galindo
- Department of Pediatric Oncology, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Boston, MA, USA.,Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Matthew Wilson
- Department of Surgery, St. Jude Children's Research Hospital, Memphis, TN, USA.,Department of Ophthalmology, Hamilton Eye Institute, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ibrahim Qaddoumi
- Department of Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
42
|
Wit JM, Oostdijk W, Losekoot M, van Duyvenvoorde HA, Ruivenkamp CAL, Kant SG. MECHANISMS IN ENDOCRINOLOGY: Novel genetic causes of short stature. Eur J Endocrinol 2016; 174:R145-73. [PMID: 26578640 DOI: 10.1530/eje-15-0937] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Accepted: 11/16/2015] [Indexed: 12/17/2022]
Abstract
The fast technological development, particularly single nucleotide polymorphism array, array-comparative genomic hybridization, and whole exome sequencing, has led to the discovery of many novel genetic causes of growth failure. In this review we discuss a selection of these, according to a diagnostic classification centred on the epiphyseal growth plate. We successively discuss disorders in hormone signalling, paracrine factors, matrix molecules, intracellular pathways, and fundamental cellular processes, followed by chromosomal aberrations including copy number variants (CNVs) and imprinting disorders associated with short stature. Many novel causes of GH deficiency (GHD) as part of combined pituitary hormone deficiency have been uncovered. The most frequent genetic causes of isolated GHD are GH1 and GHRHR defects, but several novel causes have recently been found, such as GHSR, RNPC3, and IFT172 mutations. Besides well-defined causes of GH insensitivity (GHR, STAT5B, IGFALS, IGF1 defects), disorders of NFκB signalling, STAT3 and IGF2 have recently been discovered. Heterozygous IGF1R defects are a relatively frequent cause of prenatal and postnatal growth retardation. TRHA mutations cause a syndromic form of short stature with elevated T3/T4 ratio. Disorders of signalling of various paracrine factors (FGFs, BMPs, WNTs, PTHrP/IHH, and CNP/NPR2) or genetic defects affecting cartilage extracellular matrix usually cause disproportionate short stature. Heterozygous NPR2 or SHOX defects may be found in ∼3% of short children, and also rasopathies (e.g., Noonan syndrome) can be found in children without clear syndromic appearance. Numerous other syndromes associated with short stature are caused by genetic defects in fundamental cellular processes, chromosomal abnormalities, CNVs, and imprinting disorders.
Collapse
Affiliation(s)
- Jan M Wit
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Wilma Oostdijk
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Monique Losekoot
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Hermine A van Duyvenvoorde
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Claudia A L Ruivenkamp
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| | - Sarina G Kant
- Departments of PaediatricsClinical GeneticsLeiden University Medical Center, PO Box 9600, 2300 RC Leiden, The Netherlands
| |
Collapse
|
43
|
Ossandón D, Zanolli M, López JP, Benavides F, Pérez V, Repetto GM. Molecular diagnosis in patients with retinoblastoma: Report of a series of cases. ACTA ACUST UNITED AC 2016; 91:379-84. [PMID: 27021801 DOI: 10.1016/j.oftal.2016.02.013] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 01/09/2023]
Abstract
OBJECTIVES To report the benefits of genetic diagnosis in patients with retinoblastoma. METHOD Observational study. Patients with retinoblastoma and their families were included. Demographic and clinical data were recorded. Blood and tumour samples were obtained. Next generation sequencing was performed on the samples. When deletion 13 q syndrome was suspected, cytogenetics microarray was performed (Cytoscan® HD, Affymetrix, Santa Clara, CA, USA), with a high density chip of 1.9 million of non-polymorphic probes and 750 thousand SNP probes. RESULTS Of the 7 cases were analysed 4 were male. The mean age at diagnosis was 21 months (range 5-36). Three cases had bilateral retinoblastoma, and 4 unilateral. None had family history. In all patients, blood was analysed, and a study was performed on the tissue from 2 unilateral enucleated tumours, in which 6 mutations were identified, all de novo. Just one was novel (c.164delC; case 1). One case of unilateral tumour revealed blood mosaicism, showing that his condition was inheritable, and that there is a high risk of developing retinoblastoma in the unaffected eye. The patient also has an increased risk of presenting with other primary tumours. CONCLUSION Molecular diagnosis of RB1 in patients with retinoblastoma impacts on the decision process, costs, treatment, and prognosis of patients, as well as their families.
Collapse
MESH Headings
- Child, Preschool
- Chile
- Chromosome Deletion
- Chromosome Disorders/diagnosis
- Chromosome Disorders/genetics
- Chromosomes, Human, Pair 13/genetics
- DNA Mutational Analysis
- DNA, Neoplasm/blood
- DNA, Neoplasm/genetics
- DNA, Neoplasm/isolation & purification
- Eye Neoplasms/blood
- Eye Neoplasms/chemistry
- Eye Neoplasms/diagnosis
- Eye Neoplasms/genetics
- Female
- Genes, Retinoblastoma
- Humans
- Infant
- Male
- Mosaicism
- Mutation
- Neoplasms, Multiple Primary/blood
- Neoplasms, Multiple Primary/chemistry
- Neoplasms, Multiple Primary/diagnosis
- Neoplasms, Multiple Primary/genetics
- Oligonucleotide Array Sequence Analysis
- Polymorphism, Single Nucleotide
- Retinoblastoma/blood
- Retinoblastoma/chemistry
- Retinoblastoma/diagnosis
- Retinoblastoma/genetics
- Retinoblastoma Binding Proteins/genetics
- Sequence Analysis, DNA/methods
- Ubiquitin-Protein Ligases/genetics
Collapse
Affiliation(s)
- D Ossandón
- Departamento de Oftalmología, Hospital San Juan de Dios, Santiago, Chile; Departamento de Oftalmología, Clínica Alemana de Santiago, Facultad de Medicina, Universidad del Desarrollo, Santiago, Chile
| | - M Zanolli
- Departamento de Oftalmología, Clínica Alemana de Santiago, Facultad de Medicina, Universidad del Desarrollo, Santiago, Chile.
| | - J P López
- Departamento de Oftalmología, Clínica Alemana de Santiago, Facultad de Medicina, Universidad del Desarrollo, Santiago, Chile
| | - F Benavides
- Centro de Genética y Genómica, Clínica Alemana de Santiago, Facultad de Medicina, Universidad del Desarrollo, Santiago, Chile
| | - V Pérez
- Departamento de Oncología Pediátrica (Programa PINDA), Hospital San Juan de Dios, Santiago, Chile
| | - G M Repetto
- Centro de Genética y Genómica, Clínica Alemana de Santiago, Facultad de Medicina, Universidad del Desarrollo, Santiago, Chile
| |
Collapse
|
44
|
Reyes-Palomares A, Bueno A, Rodríguez-López R, Medina MÁ, Sánchez-Jiménez F, Corpas M, Ranea JAG. Systematic identification of phenotypically enriched loci using a patient network of genomic disorders. BMC Genomics 2016; 17:232. [PMID: 26980139 PMCID: PMC4792099 DOI: 10.1186/s12864-016-2569-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Accepted: 03/07/2016] [Indexed: 11/29/2022] Open
Abstract
Background Network medicine is a promising new discipline that combines systems biology approaches and network science to understand the complexity of pathological phenotypes. Given the growing availability of personalized genomic and phenotypic profiles, network models offer a robust integrative framework for the analysis of "omics" data, allowing the characterization of the molecular aetiology of pathological processes underpinning genetic diseases. Methods Here we make use of patient genomic data to exploit different network-based analyses to study genetic and phenotypic relationships between individuals. For this method, we analyzed a dataset of structural variants and phenotypes for 6,564 patients from the DECIPHER database, which encompasses one of the most comprehensive collections of pathogenic Copy Number Variations (CNVs) and their associated ontology-controlled phenotypes. We developed a computational strategy that identifies clusters of patients in a synthetic patient network according to their genetic overlap and phenotype enrichments. Results Many of these clusters of patients represent new genotype-phenotype associations, suggesting the identification of newly discovered phenotypically enriched loci (indicative of potential novel syndromes) that are currently absent from reference genomic disorder databases such as ClinVar, OMIM or DECIPHER itself. Conclusions We provide a high-resolution map of pathogenic phenotypes associated with their respective significant genomic regions and a new powerful tool for diagnosis of currently uncharacterized mutations leading to deleterious phenotypes and syndromes. Electronic supplementary material The online version of this article (doi:10.1186/s12864-016-2569-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Armando Reyes-Palomares
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain. .,CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain. .,Present address: The European Molecular Biology Laboratory Heidelberg, 69117, Heidelberg, Germany.
| | - Aníbal Bueno
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain
| | - Rocío Rodríguez-López
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain.,CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain
| | - Miguel Ángel Medina
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain.,CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain
| | - Francisca Sánchez-Jiménez
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain.,CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain
| | - Manuel Corpas
- The Genome Analysis Centre, Norwich Research Park, Norwich, NR4 7UH, UK
| | - Juan A G Ranea
- Universidad de Málaga, Andalucía Tech, Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, and IBIMA (Biomedical Research Institute of Málaga), E-29071, Málaga, Spain. .,CIBER de Enfermedades Raras (CIBERER), E-29071, Málaga, Spain.
| |
Collapse
|
45
|
Bhavsar D, Subramanian K, Sethuraman S, Krishnan UM. Management of retinoblastoma: opportunities and challenges. Drug Deliv 2015; 23:2488-2496. [PMID: 25758593 DOI: 10.3109/10717544.2015.1016193] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Nano-delivery systems have significantly evolved over the last decade for the treatment of cancer by enabling site-specific delivery and improved bioavailability. The widely investigated nanoparticle systems are biodegradable polyesters, dendrimers, liposomes, mesoporous silica and gold nanoparticles. These particles when conjugated with different targeting motifs enhance the therapeutic efficiency of the drug molecules and biocompatibility. However, the application of such systems towards the treatment of retinoblastoma (RB), a rapidly spreading childhood eye cancer, still remains in its infancy. Nanoparticle-based systems that have been investigated for RB therapy have displayed improved drug delivery to the most restricted posterior segment of the eyes and have increased intra-vitreal half-life of the chemotherapy agents highlighting its potential in treatment of this form of cancer. This review focuses on the challenges involved in the treatment of RB and highlights the attempts made to develop nano-dimensional systems for the treatment of RB.
Collapse
Affiliation(s)
- Dhiraj Bhavsar
- a School of Chemical & Biotechnology, Centre for Nanotechnology & Advanced Biomaterials, Sastra University , Thanjavur , Tamil Nadu , India and
| | - Krishnakumar Subramanian
- b L&T Ophthalmic Pathology Department , Vision Research Foundation , Sankara Nethralaya , Chennai , Tamil Nadu , India
| | - Swaminathan Sethuraman
- a School of Chemical & Biotechnology, Centre for Nanotechnology & Advanced Biomaterials, Sastra University , Thanjavur , Tamil Nadu , India and
| | - Uma Maheswari Krishnan
- a School of Chemical & Biotechnology, Centre for Nanotechnology & Advanced Biomaterials, Sastra University , Thanjavur , Tamil Nadu , India and
| |
Collapse
|
46
|
Mitochondrial encephalomyopathy and retinoblastoma explained by compound heterozygosity of SUCLA2 point mutation and 13q14 deletion. Eur J Hum Genet 2014; 23:325-30. [PMID: 24986829 DOI: 10.1038/ejhg.2014.128] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/26/2014] [Accepted: 05/30/2014] [Indexed: 11/08/2022] Open
Abstract
Mutations in SUCLA2, encoding the ß-subunit of succinyl-CoA synthetase of Krebs cycle, are one cause of mitochondrial DNA depletion syndrome. Patients have been reported to have severe progressive childhood-onset encephalomyopathy, and methylmalonic aciduria, often leading to death in childhood. We studied two families, with children manifesting with slowly progressive mitochondrial encephalomyopathy, hearing impairment and transient methylmalonic aciduria, without mtDNA depletion. The other family also showed dominant inheritance of bilateral retinoblastoma, which coexisted with mitochondrial encephalomyopathy in one patient. We found a variant in SUCLA2 leading to Asp333Gly change, homozygous in one patient and compound heterozygous in one. The latter patient also carried a deletion of 13q14 of the other allele, discovered with molecular karyotyping. The deletion spanned both SUCLA2 and RB1 gene regions, leading to manifestation of both mitochondrial disease and retinoblastoma. We made a homology model for human succinyl-CoA synthetase and used it for structure-function analysis of all reported pathogenic mutations in SUCLA2. On the basis of our model, all previously described mutations were predicted to result in decreased amounts of incorrectly assembled protein or disruption of ADP phosphorylation, explaining the severe early lethal manifestations. However, the Asp333Gly change was predicted to reduce the activity of the otherwise functional enzyme. On the basis of our findings, SUCLA2 mutations should be analyzed in patients with slowly progressive encephalomyopathy, even in the absence of methylmalonic aciduria or mitochondrial DNA depletion. In addition, an encephalomyopathy in a patient with retinoblastoma suggests mutations affecting SUCLA2.
Collapse
|
47
|
Dehainault C, Garancher A, Castéra L, Cassoux N, Aerts I, Doz F, Desjardins L, Lumbroso L, Montes de Oca R, Almouzni G, Stoppa-Lyonnet D, Pouponnot C, Gauthier-Villars M, Houdayer C. The survival gene MED4 explains low penetrance retinoblastoma in patients with large RB1 deletion. Hum Mol Genet 2014; 23:5243-50. [PMID: 24858910 DOI: 10.1093/hmg/ddu245] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Retinoblastoma is a non-hereditary as well as an inherited pediatric tumor of the developing retina resulting from the inactivation of both copies of the RB1 tumor suppressor gene. Familial retinoblastoma is a highly penetrant genetic disease that usually develops by carrying germline mutations that inactivate one allele of the RB1 gene, leading to multiple retinoblastomas. However, large and complete germline RB1 deletions are associated with low or no tumor risk for reasons that remain unknown. In this study, we define a minimal genomic region associated with this low penetrance. This region encompasses few genes including MED4 a subunit of the mediator complex. We further show that retinoblastoma RB1 -/- cells cannot survive in the absence of MED4, both in vitro and in orthotopic xenograft models in vivo, therefore identifying MED4 as a survival gene in retinoblastoma. We propose that the contiguous loss of the adjacent retinoblastoma gene, MED4, explains the low penetrance in patients with large deletions that include both RB1 and MED4. Our findings also point to another synthetic lethal target in tumors with inactivated RB1 and highlight the importance of collateral damage in carcinogenesis.
Collapse
Affiliation(s)
| | - Alexandra Garancher
- Institut Curie, Section Recherche, Orsay, France CNRS UMR3347 INSERM U1021 Université Paris Sud and
| | | | - Nathalie Cassoux
- Département d'oncologie chirurgicale, service d'Ophtalmologie, Institut Curie, Paris, France Laboratoire d'Investigation Préclinique, Institut Curie, Paris, France
| | - Isabelle Aerts
- Département d'oncologie pédiatrique, adolescents jeunes adultes and
| | - François Doz
- Département d'oncologie pédiatrique, adolescents jeunes adultes and Université Paris Descartes, Sciences pharmaceutiques et biologiques, Sorbonne Paris Cité, Paris, France
| | - Laurence Desjardins
- Département d'oncologie chirurgicale, service d'Ophtalmologie, Institut Curie, Paris, France
| | - Livia Lumbroso
- Département d'oncologie chirurgicale, service d'Ophtalmologie, Institut Curie, Paris, France
| | | | | | - Dominique Stoppa-Lyonnet
- Service de Génétique, Institut Curie, Paris, France INSERM U830, centre de recherche de l'Institut Curie, Paris, France Université Paris Descartes, Sciences pharmaceutiques et biologiques, Sorbonne Paris Cité, Paris, France
| | - Celio Pouponnot
- Institut Curie, Section Recherche, Orsay, France CNRS UMR3347 INSERM U1021 Université Paris Sud and
| | | | - Claude Houdayer
- Service de Génétique, Institut Curie, Paris, France INSERM U830, centre de recherche de l'Institut Curie, Paris, France Université Paris Descartes, Sciences pharmaceutiques et biologiques, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
48
|
Pavone P, Briuglia S, Falsaperla R, Warm A, Pavone V, Bernardini L, Novelli A, Praticò AD, Salpietro V, Ruggieri M. Wide spectrum of congenital anomalies including choanal atresia, malformed extremities, and brain and spinal malformations in a girl with a de novo 5.6-Mb deletion of 13q12.11-13q12.13. Am J Med Genet A 2014; 164A:1734-43. [PMID: 24807585 DOI: 10.1002/ajmg.a.36391] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 11/07/2013] [Indexed: 01/31/2023]
Abstract
A 2 ½-year-old girl with multiple congenital anomalies and a de novo 5.6-Mb deletion on chromosome 13q12.11-13q12.13 is reported. She showed choanal atresia, scalp aplasia cutis, mild dysmorphic features, severe malformation of the hands and feet, Sylvian aqueductal stenosis, hydrocephalus, small cerebellum with pointed cerebellar tonsils, cervical, lumbar and sacral clefting, single central incisor and mild developmental delay. The girl's anomalies were compared with: (A) one boy reported by each of Der Kaloustian et al. [2011] and Tanteles et al. [2011] with similar, albeit smaller, 2.1 to 2.9 Mb deletions in which the abnormalities consisted of mild facial dysmorphism, mild malformations of the fingers and/or toes, and developmental delay; (B) one girl reported by Friedman et al. [2006] with similar, albeit larger, 5.7 Mb deletion with mild developmental delay and haematological abnormalities; (C) one girl reported by Slee et al. [1991] with a deletion of band q12.2 in chromosome 13, who had Moebius syndrome with facial dysmorphism, high arched palate, micrognathia, and small tongue with no abnormalities of the extremities; and (D) seven additional individuals recorded in the DECIPHER 6.0 database who all had dysmorphic features and developmental delay plus a spectrum of clinical manifestations including deafness, ataxia/oculomotor apraxia, spasticity, small testes, and mild fingers' anomalies. The deleted region hereby reported encompassed 34 known genes, including GJA3, GJB2, and GJB6, which are responsible for autosomal recessive deafness, FGF9, which plays crucial roles in embryonic neurological development, and ATP8A2, which causes a cerebellar ataxia and disequilibrium syndrome.
Collapse
Affiliation(s)
- Piero Pavone
- Unit of Pediatrics and Pediatric Emergency, University Hospital "Policlinico-Vittorio Emanuele", Catania, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Genetic testing in Tunisian families with heritable retinoblastoma using a low cost approach permits accurate risk prediction in relatives and reveals incomplete penetrance in adults. Exp Eye Res 2014; 124:48-55. [PMID: 24810223 DOI: 10.1016/j.exer.2014.04.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2014] [Revised: 04/01/2014] [Accepted: 04/18/2014] [Indexed: 01/16/2023]
Abstract
Heritable retinoblastoma is caused by oncogenic mutations in the RB1 tumor suppressor gene. Identification of these mutations in patients is important for genetic counseling and clinical management of relatives at risk. In order to lower analytical efforts, we designed a stepwise mutation detection strategy that was adapted to the spectrum of oncogenic RB1 gene mutations. We applied this strategy on 20 unrelated patients with familial and/or de novo bilateral retinoblastoma from Tunisia. In 19 (95%) patients, we detected oncogenic mutations including base substitutions, small length mutations, and large deletions. Further analyses on the origin of the mutations showed mutational mosaicism in one unilaterally affected father of a bilateral proband and incomplete penetrance in two mothers. In a large family with several retinoblastoma patients, the mutation identified in the index patient was also detected in several non-penetrant relatives. RNA analyses showed that this mutation results in an in-frame loss of exon 9. In summary, our strategy can serve as a model for RB1 mutation identification with high analytical sensitivity. Our results point out that genetic testing is needed to reveal or exclude incomplete penetrance specifically in parents of patients with sporadic disease.
Collapse
|
50
|
Jones K, Minassian BA. Genetic testing in infantile spasms identifies a chromosome 13q deletion and retinoblastoma. Pediatr Neurol 2014; 50:522-4. [PMID: 24485929 DOI: 10.1016/j.pediatrneurol.2013.11.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 11/19/2013] [Accepted: 11/23/2013] [Indexed: 11/29/2022]
Abstract
BACKGROUND Infantile spasms is an epileptic encephalopathy and the common final manifestation of numerous disparate insults to the developing brain during infancy. The varied etiologies may be structural, metabolic, genetic, or unknown. Etiological diagnosis is important as it may lead to specific therapy, which may affect developmental outcome. PATIENT We report a case of infantile spasms of unknown etiology with dysmorphic features, in which genetic copy number variation microarray testing was included in the investigation of the cause of the disease. RESULTS A large deletion of chromosome 13 was identified in the region 13q13 to 13q21.3 encompassing the retinoblastoma gene (13q14.2). Urgent ophthalmological evaluation revealed an asymptomatic retinoblastoma of the left eye, leading to early treatment. CONCLUSION This is the first case report of infantile spasms specifically associated with a chromosome 13q deletion. Chromosomal region 13q13 to 13q21.3 may contain one or more genes whose hemizygous loss leads to infantile spasms. Copy number variation testing for cryptogenic infantile spasms led to the discovery of a mutation responsible for retinoblastoma, enabling early diagnosis and treatment of a potentially life-threatening cancer. High-sensitivity molecular diagnosis improves health care and substantially reduces expenses. This shift in diagnostic evaluation is broadly relevant to health care.
Collapse
Affiliation(s)
- Kevin Jones
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.
| | - Berge A Minassian
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| |
Collapse
|