1
|
Savad S, Modarressi MH, Younesi S, Seifi-Alan M, Samadaian N, Masoomy M, Dianatpour M, Norouzi S, Amidi S, Boroumand A, Ashrafi MR, Ronagh A, Eslami M, Hashemnejad M, Nourian S, Mohammadi S, Taheri Amin MM, Heidari M, Seifi-Alan M, Shojaaldini Ardakani H, Aghamahdi F, Khalilian S, Ghafouri-Fard S. A Comprehensive Overview of NF1 Mutations in Iranian Patients. Neuromolecular Med 2024; 26:28. [PMID: 38954284 DOI: 10.1007/s12017-024-08790-5] [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: 03/16/2024] [Accepted: 04/30/2024] [Indexed: 07/04/2024]
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disorder caused by mutations in the NF1 gene. This disorder shows nearly complete penetrance and high phenotypic variability. We used the whole-exome sequencing technique to identify mutations in 32 NF1 cases from 22 Iranian families. A total of 31 variants, including 30 point mutations and one large deletion, were detected. In eight cases, variants were inherited, while they were sporadic in the remaining. Seven novel variants, including c.5576 T > G, c.6658_6659insC, c.2322dupT, c.92_93insAA, c.4360C > T, c.3814C > T, and c.4565_4566delinsC, were identified. The current study is the largest in terms of the sample size of Iranian NF1 cases with identified mutations. The results can broaden the spectrum of NF1 mutations and facilitate the process of genetic counseling in the affected families.
Collapse
Affiliation(s)
| | | | - Sarang Younesi
- Prenatal Screening Department, Nilou Laboratory, Tehran, Iran
| | - Mahnaz Seifi-Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Mona Masoomy
- Applied Biotechnology Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mehdi Dianatpour
- Stem Cell and Transgenic Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Human Genetic, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | | | - Mahmoud Reza Ashrafi
- Pediatric Neurology Division, Children's Medical Center, Pediatrics Center of Excellence, Ataxia Clinic, Tehran University of Medical Sciences, Tehran, Iran
| | - Alireza Ronagh
- Department of Pediatrics Neurologists, Shahid Bahonar Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Maryam Eslami
- Applied Biotechnology Research Center, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Department of Genetics, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Maryam Hashemnejad
- Department of Obstetrics and Gynecology, School of Medicine, Kamali Hospital, Alborz University of Medical Sciences, Karaj, Iran
| | - Shahab Nourian
- Department of Pediatrics Endocrinology and Metabolisms, Emam Ali Hospital, Alborz University of Medical Sciences and Health Services, Karaj, Iran
| | - Sanaz Mohammadi
- Comprehensive Medical Genetics Center, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Morteza Heidari
- Pediatric Neurology Division, Children's Medical Center, Pediatrics Center of Excellence, Ataxia Clinic, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahin Seifi-Alan
- Cardiovascular Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | | | - Fatemeh Aghamahdi
- Department of Pediatrics, Alborz University of Medical Sciences, Karaj, Iran
| | - Sheyda Khalilian
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
Graham JH, Schlachetzki JCM, Yang X, Breuss MW. Genomic Mosaicism of the Brain: Origin, Impact, and Utility. Neurosci Bull 2024; 40:759-776. [PMID: 37898991 PMCID: PMC11178748 DOI: 10.1007/s12264-023-01124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/16/2023] [Indexed: 10/31/2023] Open
Abstract
Genomic mosaicism describes the phenomenon where some but not all cells within a tissue harbor unique genetic mutations. Traditionally, research focused on the impact of genomic mosaicism on clinical phenotype-motivated by its involvement in cancers and overgrowth syndromes. More recently, we increasingly shifted towards the plethora of neutral mosaic variants that can act as recorders of cellular lineage and environmental exposures. Here, we summarize the current state of the field of genomic mosaicism research with a special emphasis on our current understanding of this phenomenon in brain development and homeostasis. Although the field of genomic mosaicism has a rich history, technological advances in the last decade have changed our approaches and greatly improved our knowledge. We will provide current definitions and an overview of contemporary detection approaches for genomic mosaicism. Finally, we will discuss the impact and utility of genomic mosaicism.
Collapse
Affiliation(s)
- Jared H Graham
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
| | - Xiaoxu Yang
- Department of Neurosciences, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
- Rady Children's Institute for Genomic Medicine, San Diego, 92123, CA, USA
| | - Martin W Breuss
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA.
| |
Collapse
|
3
|
Nguyen HTL, Kohl E, Bade J, Eng SE, Tosevska A, Al Shihabi A, Tebon PJ, Hong JJ, Dry S, Boutros PC, Panossian A, Gosline SJC, Soragni A. A platform for rapid patient-derived cutaneous neurofibroma organoid establishment and screening. CELL REPORTS METHODS 2024; 4:100772. [PMID: 38744290 PMCID: PMC11133839 DOI: 10.1016/j.crmeth.2024.100772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 02/10/2024] [Accepted: 04/19/2024] [Indexed: 05/16/2024]
Abstract
Localized cutaneous neurofibromas (cNFs) are benign tumors that arise in the dermis of patients affected by neurofibromatosis type 1 syndrome. cNFs are benign lesions: they do not undergo malignant transformation or metastasize. Nevertheless, they can cover a significant proportion of the body, with some individuals developing hundreds to thousands of lesions. cNFs can cause pain, itching, and disfigurement resulting in substantial socio-emotional repercussions. Currently, surgery and laser desiccation are the sole treatment options but may result in scarring and potential regrowth from incomplete removal. To identify effective systemic therapies, we introduce an approach to establish and screen cNF organoids. We optimized conditions to support the ex vivo growth of genomically diverse cNFs. Patient-derived cNF organoids closely recapitulate cellular and molecular features of parental tumors as measured by immunohistopathology, methylation, RNA sequencing, and flow cytometry. Our cNF organoid platform enables rapid screening of hundreds of compounds in a patient- and tumor-specific manner.
Collapse
Affiliation(s)
- Huyen Thi Lam Nguyen
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Emily Kohl
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jessica Bade
- Pacific Northwest National Laboratories, Seattle, WA, USA
| | - Stefan E Eng
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA
| | - Anela Tosevska
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, USA
| | - Ahmad Al Shihabi
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Peyton J Tebon
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Jenny J Hong
- Division of Hematology-Oncology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Sarah Dry
- Department of Pathology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Paul C Boutros
- Department of Human Genetics, University of California, Los Angeles, Los Angeles, CA, USA; Institute for Precision Health, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA; Department of Urology, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA
| | | | - Sara J C Gosline
- Pacific Northwest National Laboratories, Seattle, WA, USA; Department of Biomedical Engineering, Oregon Health and Sciences University, Portland, OR, USA.
| | - Alice Soragni
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, USA; Jonsson Comprehensive Cancer Center, University of California, Los Angeles, Los Angeles, CA, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
4
|
Pacot L, Masliah-Planchon J, Petcu A, Terris B, Gauthier Villars M, Lespinasse J, Wolkenstein P, Vincent-Salomon A, Vidaud D, Pasmant E. Breast cancer risk in NF1-deleted patients. J Med Genet 2024; 61:428-429. [PMID: 38154814 DOI: 10.1136/jmg-2023-109682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/05/2023] [Indexed: 12/30/2023]
Affiliation(s)
- Laurence Pacot
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, CARPEM, Paris, France
- Department of genetics, Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP.Centre-Université Paris Cité, Paris, France
| | | | - Adina Petcu
- Department of Pathology, Centre Hospitalier Métropole Savoie, Chambery, France
| | - Benoit Terris
- Department of Pathology, Hôpital Cochin, Assistance Publique-Hopitaux de Paris, Paris, France
| | | | - James Lespinasse
- Unité de Génétique médicale, Centre Hospitalier Métropole Savoie, Chambéry, France
| | - Pierre Wolkenstein
- Department of Dermatology, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, Créteil, France
- Centre d'Investigation Clinique 1430, INSERM, Hôpital Henri-Mondor, Assistance Publique-Hôpitaux de Paris, Referral Center of Neurofibromatosis, Créteil, France
| | - Anne Vincent-Salomon
- Department of Pathology, Curie Institute Hospital Group, Paris, Île-de-France, France
| | - Dominique Vidaud
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, CARPEM, Paris, France
- Department of genetics, Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP.Centre-Université Paris Cité, Paris, France
| | - Eric Pasmant
- Institut Cochin, Inserm U1016, CNRS UMR8104, Université Paris Cité, CARPEM, Paris, France
- Department of genetics, Fédération de Génétique et Médecine Génomique, Hôpital Cochin, DMU BioPhyGen, AP-HP.Centre-Université Paris Cité, Paris, France
| |
Collapse
|
5
|
Mazuelas H, Magallón-Lorenz M, Uriarte-Arrazola I, Negro A, Rosas I, Blanco I, Castellanos E, Lázaro C, Gel B, Carrió M, Serra E. Unbalancing cAMP and Ras/MAPK pathways as a therapeutic strategy for cutaneous neurofibromas. JCI Insight 2024; 9:e168826. [PMID: 38175707 PMCID: PMC11143965 DOI: 10.1172/jci.insight.168826] [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: 01/12/2023] [Accepted: 12/28/2023] [Indexed: 01/05/2024] Open
Abstract
Cutaneous neurofibromas (cNFs) are benign Schwann cell (SC) tumors arising from subepidermal glia. Individuals with neurofibromatosis type 1 (NF1) may develop thousands of cNFs, which greatly affect their quality of life. cNF growth is driven by the proliferation of NF1-/- SCs and their interaction with the NF1+/- microenvironment. We analyzed the crosstalk between human cNF-derived SCs and fibroblasts (FBs), identifying an expression signature specific to the SC-FB interaction. We validated the secretion of proteins involved in immune cell migration, suggesting a role of SC-FB crosstalk in immune cell recruitment. The signature also captured components of developmental signaling pathways, including the cAMP elevator G protein-coupled receptor 68 (GPR68). Activation of Gpr68 by ogerin in combination with the MEK inhibitor (MEKi) selumetinib reduced viability and induced differentiation and death of human cNF-derived primary SCs, a result corroborated using an induced pluripotent stem cell-derived 3D neurofibromasphere model. Similar results were obtained using other Gpr68 activators or cAMP analogs/adenylyl cyclase activators in combination with selumetinib. Interestingly, whereas primary SC cultures restarted their proliferation after treatment with selumetinib alone was stopped, the combination of ogerin-selumetinib elicited a permanent halt on SC expansion that persisted after drug removal. These results indicate that unbalancing the Ras and cAMP pathways by combining MEKi and cAMP elevators could be used as a potential treatment for cNFs.
Collapse
Affiliation(s)
- Helena Mazuelas
- Hereditary Cancer Group, Translational Cancer Research Program, and
| | | | | | - Alejandro Negro
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Barcelona, Spain
- Genetics Service, Germans Trias i Pujol University Hospital, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Inma Rosas
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Barcelona, Spain
- Genetics Service, Germans Trias i Pujol University Hospital, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Ignacio Blanco
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Barcelona, Spain
- Genetics Service, Germans Trias i Pujol University Hospital, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Elisabeth Castellanos
- Clinical Genomics Research Group, Germans Trias i Pujol Research Institute (IGTP), Can Ruti Campus, Badalona, Barcelona, Spain
- Genetics Service, Germans Trias i Pujol University Hospital, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Spain
| | - Bernat Gel
- Hereditary Cancer Group, Translational Cancer Research Program, and
- Departament de Fonaments Clínics, Facultat de Medicina i Ciències de la Salut, University of Barcelona, Barcelona, Spain
| | - Meritxell Carrió
- Hereditary Cancer Group, Translational Cancer Research Program, and
| | - Eduard Serra
- Hereditary Cancer Group, Translational Cancer Research Program, and
- Centro de Investigación Biomédica en Red de Cáncer, Spain
| |
Collapse
|
6
|
Al Zouabi L, Stefanutti M, Roumeliotis S, Le Meur G, Boumard B, Riddiford N, Rubanova N, Bohec M, Gervais L, Servant N, Bardin AJ. Molecular underpinnings and environmental drivers of loss of heterozygosity in Drosophila intestinal stem cells. Cell Rep 2023; 42:113485. [PMID: 38032794 DOI: 10.1016/j.celrep.2023.113485] [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: 08/03/2022] [Revised: 09/29/2023] [Accepted: 11/08/2023] [Indexed: 12/02/2023] Open
Abstract
During development and aging, genome mutation leading to loss of heterozygosity (LOH) can uncover recessive phenotypes within tissue compartments. This phenomenon occurs in normal human tissues and is prevalent in pathological genetic conditions and cancers. While studies in yeast have defined DNA repair mechanisms that can promote LOH, the predominant pathways and environmental triggers in somatic tissues of multicellular organisms are not well understood. Here, we investigate mechanisms underlying LOH in intestinal stem cells in Drosophila. Infection with the pathogenic bacteria, Erwinia carotovora carotovora 15, but not Pseudomonas entomophila, increases LOH frequency. Using whole genome sequencing of somatic LOH events, we demonstrate that they arise primarily via mitotic recombination. Molecular features and genetic evidence argue against a break-induced replication mechanism and instead support cross-over via double Holliday junction-based repair. This study provides a mechanistic understanding of mitotic recombination, an important mediator of LOH, and its effects on stem cells in vivo.
Collapse
Affiliation(s)
- Lara Al Zouabi
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Marine Stefanutti
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Spyridon Roumeliotis
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Gwenn Le Meur
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Benjamin Boumard
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Nick Riddiford
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Natalia Rubanova
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France; Bioinformatics, Biostatistics, Epidemiology and Computational Systems Unit, Institut Curie, PSL Research University, INSERM U900, 75005 Paris, France
| | - Mylène Bohec
- ICGex Next-Generation Sequencing Platform, Institut Curie, PSL Research University, 75005 Paris, France
| | - Louis Gervais
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France
| | - Nicolas Servant
- Bioinformatics, Biostatistics, Epidemiology and Computational Systems Unit, Institut Curie, PSL Research University, INSERM U900, 75005 Paris, France
| | - Allison J Bardin
- Genetics and Developmental Biology Department, Institut Curie, PSL Research University, Sorbonne University, CNRS UMR 3215, INSERM U934, 75248 Paris, France.
| |
Collapse
|
7
|
Coulpier F, Pulh P, Oubrou L, Naudet J, Fertitta L, Gregoire JM, Bocquet A, Schmitt AM, Wolkenstein P, Radomska KJ, Topilko P. Topical delivery of mitogen-activated protein kinase inhibitor binimetinib prevents the development of cutaneous neurofibromas in neurofibromatosis type 1 mutant mice. Transl Res 2023; 261:16-27. [PMID: 37331503 DOI: 10.1016/j.trsl.2023.06.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 05/19/2023] [Accepted: 06/13/2023] [Indexed: 06/20/2023]
Abstract
Cutaneous neurofibromas (cNFs) are a hallmark of patients with the neurofibromatosis type 1 (NF1) genetic disorder. These benign nerve sheath tumors, which can amount to thousands, develop from puberty onward, often cause pain and are considered by patients to be the primary burden of the disease. Mutations of NF1, encoding a negative regulator of the RAS signaling pathway, in the Schwann cell (SCs) lineage are considered to be at the origin of cNFs. The mechanisms governing cNFs development are poorly understood, and therapeutics to reduce cNFs are missing, mainly due to the lack of appropriate animal models. To address this, we designed the Nf1-KO mouse model that develops cNFs. Using this model, we found that cNFs development is a singular event and goes through 3 successive stages: initiation, progression, and stabilization characterized by changes in the proliferative and MAPK activities of tumor SCs. We found that skin trauma accelerated the development of cNFs and further used this model to explore the efficacy of the MEK inhibitor binimetinib to cure these tumors. We showed that while topically delivered binimetinib has a selective and minor effect on mature cNFs, the same drug prevents their development over long periods.
Collapse
Affiliation(s)
- Fanny Coulpier
- Mondor Institute for Biomedical Research, Creteil, France
| | - Pernelle Pulh
- Mondor Institute for Biomedical Research, Creteil, France
| | - Layna Oubrou
- Mondor Institute for Biomedical Research, Creteil, France
| | - Julie Naudet
- Mondor Institute for Biomedical Research, Creteil, France
| | - Laura Fertitta
- Mondor Institute for Biomedical Research, Creteil, France; Dermatology Department, Centre de Référence des Neurofibromatoses, Hôpital Henri-Mondor, AP-HP, Créteil, France
| | | | | | | | - Pierre Wolkenstein
- Mondor Institute for Biomedical Research, Creteil, France; Dermatology Department, Centre de Référence des Neurofibromatoses, Hôpital Henri-Mondor, AP-HP, Créteil, France
| | | | - Piotr Topilko
- Mondor Institute for Biomedical Research, Creteil, France.
| |
Collapse
|
8
|
Roy V, Paquet A, Touzel-Deschênes L, Khuong HT, Dupré N, Gros-Louis F. Heterozygous NF1 dermal fibroblasts modulate exosomal content to promote angiogenesis in a tissue-engineered skin model of neurofibromatosis type-1. J Neurochem 2023; 167:556-570. [PMID: 37837197 DOI: 10.1111/jnc.15982] [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: 06/15/2023] [Revised: 09/01/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023]
Abstract
Neovascularization is a critical process in tumor progression and malignant transformation associated with neurofibromatosis type 1 (NF1). Indeed, fibroblasts are known to play a key role in the tumoral microenvironment modification by producing an abundant collagenous matrix, but their contribution in paracrine communication pathways is poorly understood. Here, we hypothesized that NF1 heterozygosis in human dermal fibroblasts could promote angiogenesis through exosomes secretion. The purposes of this study are to identify the NF1 fibroblast-derived exosome protein contents and to determine their proangiogenic activity. Angiogenic proteome measurement confirmed the overexpression of VEGF and other proteins involved in vascularization. Tube formation of microvascular endothelial cells was also enhanced in presence of exosomes derived from NF1 skin fibroblasts. NF1 tissue-engineered skin (NF1-TES) generation showed a significantly denser microvessels networks compared to healthy controls. The reduction of exosomes production with an inhibitor treatment demonstrated a drastic decrease in blood vessel formation within the dermis. Our results suggest that NF1 haploinsufficiency alters the dermal fibroblast function and creates a pro-angiogenic signal via exosomes, which increases the capillary formation. This study highlights the potential of targeting exosome secretion and angiogenesis for therapeutic interventions in NF1.
Collapse
Affiliation(s)
- Vincent Roy
- Department of Surgery, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, Quebec, Canada
| | - Alexandre Paquet
- Department of Surgery, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, Quebec, Canada
| | - Lydia Touzel-Deschênes
- Department of Surgery, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, Quebec, Canada
| | - Hélène T Khuong
- Department of Surgery, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, Quebec, Canada
| | - Nicolas Dupré
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, Quebec, Canada
- Department of Neurological Sciences, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
| | - Francois Gros-Louis
- Department of Surgery, Faculty of Medicine, Laval University, Quebec City, Quebec, Canada
- Division of Regenerative Medicine, CHU de Quebec Research Center, Laval University, Quebec City, Quebec, Canada
| |
Collapse
|
9
|
Jiang C, McKay RM, Lee SY, Romo CG, Blakeley JO, Haniffa M, Serra E, Steensma MR, Largaespada D, Le LQ. Cutaneous Neurofibroma Heterogeneity: Factors that Influence Tumor Burden in Neurofibromatosis Type 1. J Invest Dermatol 2023; 143:1369-1377. [PMID: 37318402 PMCID: PMC11173230 DOI: 10.1016/j.jid.2022.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 12/01/2022] [Accepted: 12/05/2022] [Indexed: 06/16/2023]
Abstract
Neurofibromatosis type 1 is one of the most common genetic disorders of the nervous system and predisposes patients to develop benign and malignant tumors. Cutaneous neurofibromas (cNFs) are NF1-associated benign tumors that affect nearly 100% of patients with NF1. cNFs dramatically reduce patients' QOL owing to their unaesthetic appearance, physical discomfort, and corresponding psychological burden. There is currently no effective drug therapy option, and treatment is restricted to surgical removal. One of the greatest hurdles for cNF management is the variability of clinical expressivity in NF1, resulting in intrapatient and interpatient cNF tumor burden heterogeneity, that is, the variability in the presentation and evolution of these tumors. There is growing evidence that a wide array of factors are involved in the regulation of cNF heterogeneity. Understanding the mechanisms underlying this heterogeneity of cNF at the molecular, cellular, and environmental levels can facilitate the development of innovative and personalized treatment regimens.
Collapse
Affiliation(s)
- Chunhui Jiang
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Renée M McKay
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sang Y Lee
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Carlos G Romo
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Jaishri O Blakeley
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Muzlifah Haniffa
- Biosciences Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom; NIHR Newcastle Biomedical Research Center Dermatology, Newcastle University, Newcastle Upon Tyne, United Kingdom
| | - Eduard Serra
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), Barcelona, Spain
| | - Matthew R Steensma
- Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan, USA
| | - David Largaespada
- Masonic Cancer Center, University of Minnesota, Minneapolis, Minnesota, USA; Division of Hematology and Oncology, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Comprehensive Neurofibromatosis Clinic, University of Texas Southwestern Medical Center, Dallas, Texas, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA; O'Donnell Brain Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA.
| |
Collapse
|
10
|
Chen JL, Miller DT, Schmidt LS, Malkin D, Korf BR, Eng C, Kwiatkowski DJ, Giannikou K. Mosaicism in Tumor Suppressor Gene Syndromes: Prevalence, Diagnostic Strategies, and Transmission Risk. Annu Rev Genomics Hum Genet 2022; 23:331-361. [PMID: 36044908 DOI: 10.1146/annurev-genom-120121-105450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
A mosaic state arises when pathogenic variants are acquired in certain cell lineages during postzygotic development, and mosaic individuals may present with a generalized or localized phenotype. Here, we review the current state of knowledge regarding mosaicism for eight common tumor suppressor genes-NF1, NF2, TSC1, TSC2, PTEN, VHL, RB1, and TP53-and their related genetic syndromes/entities. We compare and discuss approaches for comprehensive diagnostic genetic testing, the spectrum of variant allele frequency, and disease severity. We also review affected individuals who have no mutation identified after conventional genetic analysis, as well as genotype-phenotype correlations and transmission risk for each tumor suppressor gene in full heterozygous and mosaic patients. This review provides new insight into similarities as well as marked differences regarding the appreciation of mosaicism in these tumor suppressor syndromes.
Collapse
Affiliation(s)
- Jillian L Chen
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine and Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA; .,Boston University School of Medicine, Boston, Massachusetts, USA
| | - David T Miller
- Division of Genetics and Genomics, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Laura S Schmidt
- Urologic Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, USA.,Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - David Malkin
- Division of Haematology/Oncology, The Hospital for Sick Children, Toronto, Ontario, Canada.,Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Bruce R Korf
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA.,Germline High Risk Cancer Focus Group, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, USA
| | - David J Kwiatkowski
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine and Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA;
| | - Krinio Giannikou
- Cancer Genetics Laboratory, Division of Pulmonary and Critical Care Medicine and Division of Genetics, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA; .,Division of Hematology and Oncology, Cancer and Blood Disease Institute, Children's Hospital Los Angeles, Los Angeles, California, USA;
| |
Collapse
|
11
|
Li K, Liu SJ, Wang HB, Yin CY, Huang YS, Guo WT. Schwannomatosis patient who was followed up for fifteen years: A case report. World J Clin Cases 2022; 10:6981-6990. [PMID: 36051151 PMCID: PMC9297415 DOI: 10.12998/wjcc.v10.i20.6981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 04/05/2022] [Accepted: 06/03/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Schwannomatosis is a rare disease characterized by multiple schwannomas of the whole body. Although benign, schwannomatosis that occurs in important areas of the body, such as the brain and spinal canal, can cause considerable disability and mortality. The disease is rare, frequent and relapsing, and this poses a diagnostic and therapeutic challenge.
CASE SUMMARY A 40-year-old male had multiple masses all over his body, starting at the age of 19. Four years prior, he started to experience a progressive decrease in muscle strength in both lower limbs and developed urinary and defecation dysfunctions, and gradual paralysis. One month prior, the patient developed pain and numbness in his left forearm. The patient had undergone five surgical procedures for this disease in our department. Based on the family history, imaging examinations, pathological biopsy and molecular biological examinations, the diagnosis of schwannomatosis was confirmed. This time, the patient was admitted to our hospital again for a 6th operation because of the pain and numbness in his left forearm. After the operation, the patient's symptoms improved significantly; the patient recovered and was discharged from the hospital. At the last telephone follow-up, the patient reported a poor general condition but was alive.
CONCLUSION Here, we report a rare case of schwannomatosis. We conducted 15 years of patient follow-up and treatment, and analyzed the timing of surgery and patient psychology. This case will further extend our overall understanding of the diagnosis and treatment of this rare tumor.
Collapse
Affiliation(s)
- Kai Li
- Department of Spine Surgery, The Second Hospital affiliated to Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Si-Jing Liu
- Department of Spine Surgery, The Second Hospital affiliated to Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Huai-Bo Wang
- Department of Spine Surgery, The Second Hospital affiliated to Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Chang-Yu Yin
- Department of Spine Surgery, The Second Hospital affiliated to Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Yong-Sheng Huang
- Department of Spine Surgery, The Second Hospital affiliated to Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| | - Wei-Tao Guo
- Department of Spine Surgery, The Second Hospital affiliated to Guangdong Medical University, Zhanjiang 524000, Guangdong Province, China
| |
Collapse
|
12
|
Tong S, Devine WP, Shieh JT. Tumor and Constitutional Sequencing for Neurofibromatosis Type 1. JCO Precis Oncol 2022; 6:e2100540. [PMID: 35584348 PMCID: PMC9200388 DOI: 10.1200/po.21.00540] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
NF1 variants in tumors are important to recognize, as multiple mechanisms may give rise to biallelic variants. Both deletions and copy-neutral loss of heterozygosity (LOH) are potential mechanisms of NF1 loss, distinct from point mutations, and additional genes altered may drive different tumor types. This study investigates whether tumors from individuals with neurofibromatosis type 1 (NF1) demonstrate additional gene variants and detects NF1 second hits using paired germline and somatic sequencing. In addition, rare tumor types in NF1 may also be characterized by tumor sequencing. NF1 second hits are primarily copy-neutral LOH and offer opportunity for variant interpretation
Collapse
Affiliation(s)
- Schuyler Tong
- Division of Hematology/Oncology, Pediatrics, Benioff Children's Hospital Oakland, University of California San Francisco, San Francisco, CA
| | - W Patrick Devine
- Department of Pathology, University of California San Francisco, San Francisco, CA.,Institute for Human Genetics, University of California San Francisco, San Francisco, CA
| | - Joseph T Shieh
- Institute for Human Genetics, University of California San Francisco, San Francisco, CA.,Division of Medical Genetics, Pediatrics, Benioff Children's Hospital, University of California San Francisco, San Francisco, CA
| |
Collapse
|
13
|
Garcia B, Catasus N, Ros A, Rosas I, Negro A, Guerrero-Murillo M, Valero AM, Duat-Rodriguez A, Becerra JL, Bonache S, Lázaro Garcia C, Comas C, Bielsa I, Serra E, Hernández-Chico C, Martin Y, Castellanos E, Blanco I. Neurofibromatosis type 1 families with first-degree relatives harbouring distinct NF1 pathogenic variants. Genetic counselling and familial diagnosis: what should be offered? J Med Genet 2022; 59:1017-1023. [PMID: 35121649 DOI: 10.1136/jmedgenet-2021-108301] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/09/2022] [Indexed: 11/03/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disorder caused by pathogenic variants in NF1 Recently, NF1 testing has been included as a clinical criterion for NF1 diagnosis. Additionally, preconception genetic counselling in patients with NF1 focuses on a 50% risk of transmitting the familial variant as the risk of having a sporadic NF1 is considered the same as the general population. METHODS 829 individuals, 583 NF1 sporadic cases and 246 patients with NF1 with documented family history, underwent genetic testing for NF1. Genotyping and segregation analysis of NF1 familial variants was determined by microsatellite analysis and NF1 sequencing. RESULTS The mutational analysis of NF1 in 154 families with two or more affected cases studied showed the co-occurrence of two different NF1 germline pathogenic variants in four families. The estimated mutation rate in those families was 3.89×10-3, 20 times higher than the NF1 mutation rate (~2×10-4) (p=0.0008). Furthermore, the co-occurrence of two different NF1 germline pathogenic variants in these families was 1:39, 60 times the frequency of sporadic NF1 (1:2500) (p=0.003). In all cases, the de novo NF1 pathogenic variant was present in a descendant of an affected male. In two cases, variants were detected in the inherited paternal wild-type allele. CONCLUSIONS Our results, together with previous cases reported, suggest that the offspring of male patients with NF1 could have an increased risk of experiencing de novo NF1 pathogenic variants. This observation, if confirmed in additional cohorts, could have relevant implications for NF1 genetic counselling, family planning and NF1 genetic testing.
Collapse
Affiliation(s)
- Belen Garcia
- Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain
| | - Nuria Catasus
- Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain
| | - Andrea Ros
- Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain
| | - Inma Rosas
- Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Unit-Genetics Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alejandro Negro
- Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Unit-Genetics Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Mercedes Guerrero-Murillo
- Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Unit-Genetics Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Ana Maria Valero
- Servicio de Genética, Hospital Universitario Ramon y Cajal, Madrid, Spain
| | - Anna Duat-Rodriguez
- Neurology Service, Hospital Infantil Universitario Niño Jesús, Madrid, Spain
| | - Juan Luis Becerra
- Neurology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Sandra Bonache
- Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Unit-Genetics Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Conxi Lázaro Garcia
- Hereditary Cancer Program, Program in Molecular Mechanisms and Experimental Therapy in Oncology (Oncobell), IDIBELL, Catalan Institute of Oncology, L'Hospitalet de Llobregat, Spain.,Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Carmina Comas
- Department of Obstetrics, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Isabel Bielsa
- Dermatology, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Eduard Serra
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.,Hereditary Cancer Group, Fundació Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Concepción Hernández-Chico
- Servicio de Genética, IRYCIS, Hospital Universitario Ramon y Cajal, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Yolanda Martin
- Servicio de Genética, IRYCIS, Hospital Universitario Ramon y Cajal, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras, CIBERER, Valencia, Spain
| | - Elisabeth Castellanos
- Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain .,Clinical Genomics Unit-Genetics Service, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Ignacio Blanco
- Genetic Counseling Unit, Clinical Genetics Service, Northern Metropolitan Clinical Laboratory, Hospital Universitari Germans Trias i Pujol, Badalona, Spain.,Clinical Genomics Research Unit, Foundation Institute of Research in Health Sciences Germans Trias i Pujol, Badalona, Spain
| |
Collapse
|
14
|
Benatti HR, Gray-Edwards HL. Adeno-Associated Virus Delivery Limitations for Neurological Indications. Hum Gene Ther 2022; 33:1-7. [PMID: 35049369 DOI: 10.1089/hum.2022.29196.hrb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Hector Ribeiro Benatti
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| | - Heather L Gray-Edwards
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA.,Department of Radiology, University of Massachusetts Chan Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
15
|
Deb BK, Bateup HS. Modeling Somatic Mutations Associated With Neurodevelopmental Disorders in Human Brain Organoids. Front Mol Neurosci 2022; 14:787243. [PMID: 35058746 PMCID: PMC8764387 DOI: 10.3389/fnmol.2021.787243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Neurodevelopmental disorders (NDDs) are a collection of diseases with early life onset that often present with developmental delay, cognitive deficits, and behavioral conditions. In some cases, severe outcomes such as brain malformations and intractable epilepsy can occur. The mutations underlying NDDs may be inherited or de novo, can be gain- or loss-of-function, and can affect one or more genes. Recent evidence indicates that brain somatic mutations contribute to several NDDs, in particular malformations of cortical development. While advances in sequencing technologies have enabled the detection of these somatic mutations, the mechanisms by which they alter brain development and function are not well understood due to limited model systems that recapitulate these events. Human brain organoids have emerged as powerful models to study the early developmental events of the human brain. Brain organoids capture the developmental progression of the human brain and contain human-enriched progenitor cell types. Advances in human stem cell and genome engineering provide an opportunity to model NDD-associated somatic mutations in brain organoids. These organoids can be tracked throughout development to understand the impact of somatic mutations on early human brain development and function. In this review, we discuss recent evidence that somatic mutations occur in the developing human brain, that they can lead to NDDs, and discuss how they could be modeled using human brain organoids.
Collapse
Affiliation(s)
- Bipan K. Deb
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
| | - Helen S. Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, United States
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, United States
- Chan Zuckerberg Biohub, San Francisco, CA, United States
- *Correspondence: Helen S. Bateup
| |
Collapse
|
16
|
Implications of mosaicism in variant interpretation: A case of a de novo homozygous NF1 variant. Eur J Med Genet 2021; 64:104236. [PMID: 33965620 DOI: 10.1016/j.ejmg.2021.104236] [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: 11/08/2020] [Revised: 02/21/2021] [Accepted: 04/28/2021] [Indexed: 11/23/2022]
Abstract
Neurofibromatosis type 1 is a common multisystem autosomal dominant syndrome caused by pathogenic heterozygous variants in the neurofibromin gene (NF1). It is associated with a substantially increased cancer risk. Mosaicism for NF1 has been clinically well-established for "second hit" variants in skin lesions and tumor tissues. Here, we report on a 3-month-old boy with multiple café au lait macules (CAMs) and juvenile myelomonocytic leukemia (JMML) who was found to carry a previously established pathogenic NF1 variant (c.586+5G>A), as revealed by whole-exome sequencing. Surprisingly, however, this variant was detected in the homozygous state in the patient and was absent in the parents and siblings. Deep sequencing of this variant using blood, buccal swabs and skin samples was performed. As expected for an NF1 gene mutation promoting JMML, the variant was detected in 90.6% of the blood DNA reads, in sharp contrast to the mere 5% and 0.74% of reads in the saliva- and skin fibroblast-derived DNA, respectively. Our analysis, therefore, confirmed postzygotic origin of the variant followed by a mitotic event resulting in its homozygosity, although we could not differentiate between the possibilities of a gene conversion and mitotic crossover. Apparently de novo homozygous variants should trigger a careful investigation into mosaicism to achieve accurate interpretation.
Collapse
|
17
|
Modeling tumors of the peripheral nervous system associated with Neurofibromatosis type 1: Reprogramming plexiform neurofibroma cells. Stem Cell Res 2020; 49:102068. [PMID: 33160273 DOI: 10.1016/j.scr.2020.102068] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 10/16/2020] [Accepted: 10/20/2020] [Indexed: 11/24/2022] Open
Abstract
Plexiform neurofibromas (pNFs) are benign tumors of the peripheral nervous system (PNS) that can progress towards a deadly soft tissue sarcoma termed malignant peripheral nerve sheath tumor (MPNST). pNFs appear during development in the context of the genetic disease Neurofibromatosis type 1 (NF1) due to the complete loss of the NF1 tumor suppressor gene in a cell of the neural crest (NC) - Schwann cell (SC) axis of differentiation. NF1(-/-) cells from pNFs can be reprogrammed into induced pluripotent stem cells (iPSCs) that exhibit an increased proliferation rate and maintain full iPSC properties. Efficient protocols for iPSC differentiation towards NC and SC exist and thus NC cells can be efficiently obtained from NF1(-/-) iPSCs and further differentiated towards SCs. In this review, we will focus on the iPSC modeling of pNFs, including the reprogramming of primary pNF-derived cells, the properties of pNF-derived iPSCs, the capacity to differentiate towards the NC-SC lineage, and how well iPSC-derived NF1(-/-) SC spheroids recapitulate pNF-derived primary SCs. The potential uses of NF1(-/-) iPSCs in pNF modeling and a future outlook are discussed.
Collapse
|
18
|
Abstract
Mosaicism refers to the occurrence of two or more genomes in an individual derived from a single zygote. Germline mosaicism is a mutation that is limited to the gonads and can be transmitted to offspring. Somatic mosaicism is a postzygotic mutation that occurs in the soma, and it may occur at any developmental stage or in adult tissues. Mosaic variation may be classified in six ways: (a) germline or somatic origin, (b) class of DNA mutation (ranging in scale from single base pairs to multiple chromosomes), (c) developmental context, (d) body location(s), (e) functional consequence (including deleterious, neutral, or advantageous), and (f) additional sources of mosaicism, including mitochondrial heteroplasmy, exogenous DNA sources such as vectors, and epigenetic changes such as imprinting and X-chromosome inactivation. Technological advances, including single-cell and other next-generation sequencing, have facilitated improved sensitivity and specificity to detect mosaicism in a variety of biological contexts.
Collapse
Affiliation(s)
- Jeremy Thorpe
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland 21205, USA; , .,Program in Biochemistry, Cellular, and Molecular Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, USA;
| | - Ikeoluwa A Osei-Owusu
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland 21205, USA; , .,Program in Human Genetics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA;
| | | | - Rossella Tupler
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Medical School, Worcester, Massachusetts 01605, USA.,Department of Biomedical, Metabolic, and Neural Sciences, University of Modena and Reggio Emilia, 41125 Modena, Italy;
| | - Jonathan Pevsner
- Department of Neurology, Kennedy Krieger Institute, Baltimore, Maryland 21205, USA; , .,Program in Biochemistry, Cellular, and Molecular Biology, Johns Hopkins School of Medicine, Baltimore, Maryland 21287, USA; .,Program in Human Genetics, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA; .,Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, Maryland 21205, USA
| |
Collapse
|
19
|
Roy V, Magne B, Vaillancourt-Audet M, Blais M, Chabaud S, Grammond E, Piquet L, Fradette J, Laverdière I, Moulin VJ, Landreville S, Germain L, Auger FA, Gros-Louis F, Bolduc S. Human Organ-Specific 3D Cancer Models Produced by the Stromal Self-Assembly Method of Tissue Engineering for the Study of Solid Tumors. BIOMED RESEARCH INTERNATIONAL 2020; 2020:6051210. [PMID: 32352002 PMCID: PMC7178531 DOI: 10.1155/2020/6051210] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2019] [Revised: 02/07/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Cancer research has considerably progressed with the improvement of in vitro study models, helping to understand the key role of the tumor microenvironment in cancer development and progression. Over the last few years, complex 3D human cell culture systems have gained much popularity over in vivo models, as they accurately mimic the tumor microenvironment and allow high-throughput drug screening. Of particular interest, in vitrohuman 3D tissue constructs, produced by the self-assembly method of tissue engineering, have been successfully used to model the tumor microenvironment and now represent a very promising approach to further develop diverse cancer models. In this review, we describe the importance of the tumor microenvironment and present the existing in vitro cancer models generated through the self-assembly method of tissue engineering. Lastly, we highlight the relevance of this approach to mimic various and complex tumors, including basal cell carcinoma, cutaneous neurofibroma, skin melanoma, bladder cancer, and uveal melanoma.
Collapse
Affiliation(s)
- Vincent Roy
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Brice Magne
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Maude Vaillancourt-Audet
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Mathieu Blais
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Stéphane Chabaud
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Emil Grammond
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
| | - Léo Piquet
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
| | - Julie Fradette
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Isabelle Laverdière
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
- Faculty of Pharmacy, Université Laval and CHU de Québec-Université Laval Research Center, Oncology Division, Québec, QC, Canada
| | - Véronique J. Moulin
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Solange Landreville
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Centre de Recherche sur le Cancer de l'Université Laval, Québec, QC, Canada
- Department of Ophthalmology, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Lucie Germain
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François A. Auger
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François Gros-Louis
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - Stéphane Bolduc
- Centre de Recherche du CHU de Québec-Université Laval, Axe Médecine Régénératrice, Québec, QC, Canada
- Centre de Recherche en Organogénèse Expérimentale de l'Université Laval/LOEX, Québec, QC, Canada
- Department of Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| |
Collapse
|
20
|
Hozumi K, Fukuoka H, Odake Y, Takeuchi T, Uehara T, Sato T, Inoshita N, Yoshida K, Matsumoto R, Bando H, Hirota Y, Iguchi G, Taniguchi M, Otsuki N, Nishigori C, Kosaki K, Hasegawa T, Ogawa W, Takahashi Y. Acromegaly caused by a somatotroph adenoma in patient with neurofibromatosis type 1. Endocr J 2019; 66:853-857. [PMID: 31189769 DOI: 10.1507/endocrj.ej19-0035] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Although acromegaly has been reported in patients with Neurofibromatosis type 1 (NF1), these cases have not been associated with growth hormone (GH)-producing somatotroph adenoma, but with optic pathway glioma. A 68 year-old Japanese woman, who had been clinically diagnosed with NF1, was referred to our hospital due to a thyroid tumor and hypercalcemia. Acromegaly was suspected due to her facial features, and subsequent examinations revealed the presence of GH excess with a pituitary tumor, leading to the diagnosis of acromegaly. Histological and immunohistochemical analysis demonstrated an eosinophilic pituitary adenoma with diffuse positivity for GH, indicating typical somatotroph adenoma. In addition, her thyroid tumor was diagnosed histologically as follicular thyroid carcinoma (FTC) with primary hyperparathyroidism (PHPT). To investigate the pathogenesis of this untypical multiple endocrine tumor case of NF1, genetic analysis was performed using peripheral leukocytes and tissue of resected tumors. A heterozygous novel germline nonsense mutation (p.Arg1534*) in exon 35 of the NF1 gene was detected from peripheral leukocytes, which results in a truncated protein lacking the critical domain for GTPase activity, strongly suggesting its causal role in NF1. The loss of heterozygosity (LOH) in exon 35 of the NF1 gene was not detected in the somatotroph adenoma, parathyroid adenoma, and FTC. Although any mutations of the following genes; MEN1, CDKN1B, and PAX8-PPARγ were not detected, a heterozygous GNAS R201C mutation was detected in the somatotroph adenoma. To our knowledge, this is the first rare MEN1-like case of genetically diagnosed NF1 complicated with acromegaly caused by a somatotroph adenoma.
Collapse
Affiliation(s)
- Kaori Hozumi
- Division of Diabetes and Endocrinology, Kobe University Hospital, Hyogo 650-8511, Japan
| | - Hidenori Fukuoka
- Division of Diabetes and Endocrinology, Kobe University Hospital, Hyogo 650-8511, Japan
| | - Yukiko Odake
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Takehito Takeuchi
- Division of Diabetes and Endocrinology, Kobe University Hospital, Hyogo 650-8511, Japan
| | - Tomoko Uehara
- Center for Medical Genetics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Takeshi Sato
- Department of Pediatrics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Naoko Inoshita
- Department of Pathology, Toranomon Hospital, Tokyo 105-8470, Japan
| | - Kenichi Yoshida
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Ryusaku Matsumoto
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Hironori Bando
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Yushi Hirota
- Division of Diabetes and Endocrinology, Kobe University Hospital, Hyogo 650-8511, Japan
| | - Genzo Iguchi
- Division of Diabetes and Endocrinology, Kobe University Hospital, Hyogo 650-8511, Japan
| | - Masaaki Taniguchi
- Department of Neurosurgery, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Naoki Otsuki
- Department of Otolaryngology-Head and Neck Surgery, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Chikako Nishigori
- Division of Dermatology, Department of Internal Related, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Tomonobu Hasegawa
- Department of Pediatrics, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Wataru Ogawa
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| | - Yutaka Takahashi
- Division of Diabetes and Endocrinology, Kobe University Graduate School of Medicine, Hyogo 650-8511, Japan
| |
Collapse
|
21
|
Trevisson E, Morbidoni V, Forzan M, Daolio C, Fumini V, Parrozzani R, Cassina M, Midena E, Salviati L, Clementi M. The Arg1038Gly missense variant in the NF1 gene causes a mild phenotype without neurofibromas. Mol Genet Genomic Med 2019; 7:e616. [PMID: 30843352 PMCID: PMC6503065 DOI: 10.1002/mgg3.616] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2018] [Revised: 01/16/2019] [Accepted: 02/11/2019] [Indexed: 01/08/2023] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is an autosomal dominant condition caused by inactivating mutations of the NF1 gene. The wide allelic heterogeneity of this condition, with more than 3,000 pathogenic variants reported so far, is paralleled by its high clinical variability, which is observed even within the same family. The definition of genotype–phenotype correlations has been hampered by the complexity of the NF1 gene and, although a few exceptions have been recognized, the clinical course remains unpredictable in most patients. Methods Sequencing of NF1 in patients with cafè‐au‐lait spots identified the c.3112A>G variant. RNA analysis and a minigene assay were employed to investigate splicing. Results Here we report a novel genotype–phenotype correlation in NF1: the identification of the missense variant NM_000267.3:c.3112A>G p.(Arg1038Gly) in seven individuals from two unrelated families with a mild phenotype. All the patients manifest cafè‐au‐lait spots without neurofibromas or other NF1–associated complications, and Noonan syndrome features in most cases. The missense variant was not previously reported in available databases, segregates with the phenotype and involves a highly conserved residue. Both a minigene assay and patient's RNA analysis excluded an effect on splicing. Conclusion Our data support the correlation of the p.Arg1038Gly missense substitution with the cutaneous phenotype without neurofibromas or other complications. This finding may have relevant implications for patients and genetic counseling, but also to get insights into the function of neurofibromin.
Collapse
Affiliation(s)
- Eva Trevisson
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy.,Laboratorio di Genetica Clinica ed Epidemiologica, Istituto di Ricerca Pediatrica, IRP, Padova, Italy
| | - Valeria Morbidoni
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy.,Laboratorio di Genetica Clinica ed Epidemiologica, Istituto di Ricerca Pediatrica, IRP, Padova, Italy
| | - Monica Forzan
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy.,Laboratorio di Genetica Clinica ed Epidemiologica, Istituto di Ricerca Pediatrica, IRP, Padova, Italy
| | | | - Valentina Fumini
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy
| | | | - Matteo Cassina
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy
| | - Edoardo Midena
- Department of Neurosciences, University of Padova, Padova, Italy.,IRCCS-Fondazione Bietti, Rome, Italy
| | - Leonardo Salviati
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy.,Laboratorio di Genetica Clinica ed Epidemiologica, Istituto di Ricerca Pediatrica, IRP, Padova, Italy
| | - Maurizio Clementi
- Department of Women's and Children's Health, Clinical Genetics Unit, University of Padova, Padova, Italy.,Laboratorio di Genetica Clinica ed Epidemiologica, Istituto di Ricerca Pediatrica, IRP, Padova, Italy
| |
Collapse
|
22
|
Reprogramming Captures the Genetic and Tumorigenic Properties of Neurofibromatosis Type 1 Plexiform Neurofibromas. Stem Cell Reports 2019; 12:411-426. [PMID: 30713041 PMCID: PMC6373434 DOI: 10.1016/j.stemcr.2019.01.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 12/31/2018] [Accepted: 01/01/2019] [Indexed: 12/20/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a tumor predisposition genetic disease caused by mutations in the NF1 tumor suppressor gene. Plexiform neurofibromas (PNFs) are benign Schwann cell (SC) tumors of the peripheral nerve sheath that develop through NF1 inactivation and can progress toward a malignant soft tissue sarcoma. There is a lack of non-perishable model systems to investigate PNF development. We reprogrammed PNF-derived NF1(-/-) cells, descendants from the tumor originating cell. These NF1(-/-)-induced pluripotent stem cells (iPSCs) captured the genomic status of PNFs and were able to differentiate toward neural crest stem cells and further to SCs. iPSC-derived NF1(-/-) SCs exhibited a continuous high proliferation rate, poor myelination ability, and a tendency to form 3D spheres that expressed the same markers as their PNF-derived primary SC counterparts. They represent a valuable model to study and treat PNFs. PNF-derived iPSC lines were banked for making them available.
Collapse
|
23
|
Somatic mosaicism and neurodevelopmental disease. Nat Neurosci 2018; 21:1504-1514. [PMID: 30349109 DOI: 10.1038/s41593-018-0257-3] [Citation(s) in RCA: 156] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 09/21/2018] [Indexed: 12/19/2022]
Abstract
Traditionally, we have considered genetic mutations that cause neurodevelopmental diseases to be inherited or de novo germline mutations. Recently, we have come to appreciate the importance of de novo somatic mutations, which occur postzygotically and are thus present in only a subset of the cells of an affected individual. The advent of next-generation sequencing and single-cell sequencing technologies has shown that somatic mutations contribute to normal and abnormal human brain development. Somatic mutations are one important cause of neuronal migration and brain overgrowth disorders, as suggested by visible focal lesions. In addition, somatic mutations contribute to neurodevelopmental diseases without visible lesions, including epileptic encephalopathies, intellectual disability, and autism spectrum disorder, and may contribute to a broad range of neuropsychiatric diseases. Studying somatic mutations provides insight into the mechanisms underlying human brain development and neurodevelopmental diseases and has important implications for diagnosis and treatment.
Collapse
|
24
|
Isakson SH, Rizzardi AE, Coutts AW, Carlson DF, Kirstein MN, Fisher J, Vitte J, Williams KB, Pluhar GE, Dahiya S, Widemann BC, Dombi E, Rizvi T, Ratner N, Messiaen L, Stemmer-Rachamimov AO, Fahrenkrug SC, Gutmann DH, Giovannini M, Moertel CL, Largaespada DA, Watson AL. Genetically engineered minipigs model the major clinical features of human neurofibromatosis type 1. Commun Biol 2018; 1:158. [PMID: 30302402 PMCID: PMC6168575 DOI: 10.1038/s42003-018-0163-y] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 09/07/2018] [Indexed: 12/13/2022] Open
Abstract
Neurofibromatosis Type 1 (NF1) is a genetic disease caused by mutations in Neurofibromin 1 (NF1). NF1 patients present with a variety of clinical manifestations and are predisposed to cancer development. Many NF1 animal models have been developed, yet none display the spectrum of disease seen in patients and the translational impact of these models has been limited. We describe a minipig model that exhibits clinical hallmarks of NF1, including café au lait macules, neurofibromas, and optic pathway glioma. Spontaneous loss of heterozygosity is observed in this model, a phenomenon also described in NF1 patients. Oral administration of a mitogen-activated protein kinase/extracellular signal-regulated kinase inhibitor suppresses Ras signaling. To our knowledge, this model provides an unprecedented opportunity to study the complex biology and natural history of NF1 and could prove indispensable for development of imaging methods, biomarkers, and evaluation of safety and efficacy of NF1-targeted therapies.
Collapse
Affiliation(s)
- Sara H Isakson
- Masonic Cancer Center, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Anthony E Rizzardi
- Recombinetics Inc., 1246 University Avenue W., Suite 301, St. Paul, MN, 55104, USA
| | - Alexander W Coutts
- Recombinetics Inc., 1246 University Avenue W., Suite 301, St. Paul, MN, 55104, USA
| | - Daniel F Carlson
- Recombinetics Inc., 1246 University Avenue W., Suite 301, St. Paul, MN, 55104, USA
| | - Mark N Kirstein
- Masonic Cancer Center, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA.,Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Room 459, 717 Delaware Street SE, Minneapolis, MN, 55414, USA
| | - James Fisher
- Department of Experimental and Clinical Pharmacology, College of Pharmacy, University of Minnesota, Room 459, 717 Delaware Street SE, Minneapolis, MN, 55414, USA
| | - Jeremie Vitte
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, 675 Charles E Young Drive S, MRL Room 2240, Los Angeles, CA, 90095, USA
| | - Kyle B Williams
- Masonic Cancer Center, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - G Elizabeth Pluhar
- Masonic Cancer Center, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA.,Department of Veterinary Clinical Sciences, College of Veterinary Medicine, University of Minnesota, 1365 Gortner Avenue, St. Paul, MN, 55108, USA
| | - Sonika Dahiya
- Division of Neuropathology, Department of Pathology and Immunology, Washington University School of Medicine, 660S. Euclid Avenue, CB 8118, St. Louis, MO, 63110, USA
| | - Brigitte C Widemann
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, CRC 1-5750, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Eva Dombi
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, CRC 1-5750, 10 Center Drive, Bethesda, MD, 20892, USA
| | - Tilat Rizvi
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati, 3333 Burnet Avenue, ML 7013, Cincinnati, OH, 45229, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Department of Pediatrics, Cincinnati Children's Hospital, University of Cincinnati, 3333 Burnet Avenue, ML 7013, Cincinnati, OH, 45229, USA
| | - Ludwine Messiaen
- Medical Genomics Laboratory, Department of Genetics, University of Alabama at Birmingham, Kaul Building, 720 20th Street South, Birmingham, AL, 35294, USA
| | - Anat O Stemmer-Rachamimov
- Department of Pathology, Massachusetts General Hospital, Warren Building, Room 333A, 55 Fruit Street, Boston, MA, 02114, USA
| | - Scott C Fahrenkrug
- Recombinetics Inc., 1246 University Avenue W., Suite 301, St. Paul, MN, 55104, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, Box 8111, 660S. Euclid Avenue, St. Louis, MO, 63110, USA
| | - Marco Giovannini
- Department of Head and Neck Surgery, David Geffen School of Medicine at UCLA and Jonsson Comprehensive Cancer Center (JCCC), University of California Los Angeles, 675 Charles E Young Drive S, MRL Room 2240, Los Angeles, CA, 90095, USA
| | - Christopher L Moertel
- Masonic Cancer Center, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA.,Department of Pediatrics, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - David A Largaespada
- Masonic Cancer Center, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA.,Department of Pediatrics, University of Minnesota, Room 3-129, Cancer Cardiovascular Research Building, 2231 6th Street SE, Minneapolis, MN, 55455, USA
| | - Adrienne L Watson
- Recombinetics Inc., 1246 University Avenue W., Suite 301, St. Paul, MN, 55104, USA.
| |
Collapse
|
25
|
Ferrer M, Gosline SJC, Stathis M, Zhang X, Guo X, Guha R, Ryman DA, Wallace MR, Kasch-Semenza L, Hao H, Ingersoll R, Mohr D, Thomas C, Verma S, Guinney J, Blakeley JO. Pharmacological and genomic profiling of neurofibromatosis type 1 plexiform neurofibroma-derived schwann cells. Sci Data 2018; 5:180106. [PMID: 29893754 PMCID: PMC5996849 DOI: 10.1038/sdata.2018.106] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/27/2018] [Indexed: 01/31/2023] Open
Abstract
Neurofibromatosis type I (NF1) is an autosomal dominant genetic condition characterized by peripheral nervous system tumors (PNSTs), including plexiform neurofibromas (pNFs) that cause nerve dysfunction, deformity, pain damage to adjacent structures, and can undergo malignant transformation. There are no effective therapies to prevent or treat pNFs. Drug discovery efforts are slowed by the ‘benign’ nature of the Schwann cells that are the progenitor cells of pNF. In this work we characterize a set of pNF-derived cell lines at the genomic level (via SNP Arrays, RNAseq, and Whole Exome- Sequencing), and carry out dose response-based quantitative high-throughput screening (qHTS) with a collection of 1,912 oncology-focused compounds in a 1536-well microplate cell proliferation assays. Through the characterization and screening of NF1−/−, NF1+/+ and NF1+/− Schwann cell lines, this resource introduces novel therapeutic avenues for the development for NF1 associated pNF as well as all solid tumors with NF1 somatic mutations. The integrated data sets are openly available for further analysis at http://www.synapse.org/pnfCellCulture.
Collapse
Affiliation(s)
- Marc Ferrer
- National Center for Advancing Translational Sciences (NCATS), Division of Pre-clinical Innovation, National Institutes of Health, Bethesda, MD, USA
| | | | - Marigo Stathis
- Neurofibromatosis Therapeutic Acceleration Program (NTAP), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiaohu Zhang
- National Center for Advancing Translational Sciences (NCATS), Division of Pre-clinical Innovation, National Institutes of Health, Bethesda, MD, USA
| | | | - Rajarshi Guha
- National Center for Advancing Translational Sciences (NCATS), Division of Pre-clinical Innovation, National Institutes of Health, Bethesda, MD, USA
| | - Dannielle A Ryman
- Neurofibromatosis Therapeutic Acceleration Program (NTAP), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Margaret R Wallace
- Department of Molecular Genetics & Microbiology, University of Florida College of Medicine, Gainesville, FL, USA
| | - Laura Kasch-Semenza
- Genetic Resources Core Facility (GRCF), Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Haiping Hao
- Deep Sequencing & Microarray Core, Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Roxann Ingersoll
- Genetic Resources Core Facility (GRCF), Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - David Mohr
- Center for Inherited Disease Research (CIDR), Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Craig Thomas
- National Center for Advancing Translational Sciences (NCATS), Division of Pre-clinical Innovation, National Institutes of Health, Bethesda, MD, USA
| | - Sharad Verma
- Neurofibromatosis Therapeutic Acceleration Program (NTAP), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jaishri O Blakeley
- Neurofibromatosis Therapeutic Acceleration Program (NTAP), Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
26
|
Carrió M, Gel B, Terribas E, Zucchiatti AC, Moliné T, Rosas I, Teulé Á, Ramón Y Cajal S, López-Gutiérrez JC, Blanco I, Castellanos E, Lázaro C, Stemmer-Rachamimov A, Romagosa C, Serra E. Analysis of intratumor heterogeneity in Neurofibromatosis type 1 plexiform neurofibromas and neurofibromas with atypical features: Correlating histological and genomic findings. Hum Mutat 2018; 39:1112-1125. [PMID: 29774626 DOI: 10.1002/humu.23552] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 04/29/2018] [Accepted: 05/12/2018] [Indexed: 01/11/2023]
Abstract
Plexiform neurofibromas (PNFs) are benign peripheral nerve sheath tumors involving large nerves present in 30%-50% Neurofibromatosis type 1 (NF1) patients. Atypical neurofibromas (ANF) are distinct nodular lesions with atypical features on histology that arise from PNFs. The risk and timeline of malignant transformation in ANF is difficult to assess. A recent NIH workshop has stratified ANFs and separated a subgroup with multiple atypical features and higher risk of malignant transformation termed atypical neurofibromatous neoplasms with uncertain biological potential (ANNUBP). We performed an analysis of intratumor heterogeneity on eight PNFs to link histological and genomic findings. Tumors were homogeneous although histological and molecular heterogeneity was identified. All tumors were 2n, almost mutation-free and had a clonal NF1(-/-) origin. Two ANFs from the same patient showed atypical features on histology and deletions of CDKN2A/B. One of the ANFs exhibited different areas in which the degree of histological atypia correlated with the heterozygous or homozygous loss of the CDKN2A/B loci. CDKN2A/B deletions in different areas originated independently. Results may indicate that loss of a single CDKN2A/B copy in NF1(-/-) cells is sufficient to start ANF development and that total inactivation of both copies of CDKN2A/B is necessary to form an ANNUBP.
Collapse
Affiliation(s)
- Meritxell Carrió
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), PMPPC-CIBERONC, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Bernat Gel
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), PMPPC-CIBERONC, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Ernest Terribas
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), PMPPC-CIBERONC, Can Ruti Campus, Badalona, Barcelona, Spain
| | | | - Teresa Moliné
- Department of Pathology, Vall d'Hebron University Hospital (VHIR-CIBERONC), Barcelona, Spain
| | - Inma Rosas
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), PMPPC-CIBERONC, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Álex Teulé
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL-CIBERONC), L'Hospitalet de Llobregat, Barcelona, Spain
| | - Santiago Ramón Y Cajal
- Department of Pathology, Vall d'Hebron University Hospital (VHIR-CIBERONC), Barcelona, Spain
| | | | - Ignacio Blanco
- Clinical Genetics and Genetic Counselling Program, Germans Trias i Pujol University Hospital (HUGTiP), Can Ruti Campus, Badalona, Barcelona, Spain
| | - Elisabeth Castellanos
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), PMPPC-CIBERONC, Can Ruti Campus, Badalona, Barcelona, Spain
| | - Conxi Lázaro
- Hereditary Cancer Program, Catalan Institute of Oncology (ICO-IDIBELL-CIBERONC), L'Hospitalet de Llobregat, Barcelona, Spain
| | | | - Cleofé Romagosa
- Department of Pathology, Vall d'Hebron University Hospital (VHIR-CIBERONC), Barcelona, Spain
| | - Eduard Serra
- Hereditary Cancer Group, Germans Trias i Pujol Research Institute (IGTP), PMPPC-CIBERONC, Can Ruti Campus, Badalona, Barcelona, Spain
| |
Collapse
|
27
|
Huang L, Wu X, Ding Y, Qi L, Li W, Huang G, Dai M, Zhang B. Recurrent multiple neurofibromatosis type 1 of the right lower limb. DER ORTHOPADE 2018. [PMID: 29520415 DOI: 10.1007/s00132-017-3518-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Neurofibromatosis type 1 is an autosomal dominant inherited disease, which is characterized by the presence of multiple neurofibromas. We encountered a case in which a sporadic dispersed neurofibroma recurred locally on numerous occasions extending over 16 years. The patient developed multiple masses with a focus of neurofibroma on the right lower limb, which were excised. The patient was initially diagnosed with inflammatory changes via computed tomography and magnetic resonance imaging; however, subsequently, pathological and immunohistochemical examinations revealed an intraneural neurofibroma. The patient underwent a comprehensive and complete local resection several times. After a continuous postoperative follow-up strategy, the patient recovered well. This report describes a case of primary manifestations of multiple and recurrent neurofibromas. We aim to emphasize the possibility of a unique, recurrent, non-healing neurofibroma and review the diagnostic techniques utilized to reach a definitive diagnosis. Early and complete surgical resection is an effective method to treat and prevent this type of neurofibroma.
Collapse
Affiliation(s)
- Leitao Huang
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Xia Wu
- Department of College of pharmacy, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Yi Ding
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Lai Qi
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Wei Li
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Gendong Huang
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China
| | - Min Dai
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China.
| | - Bin Zhang
- Department of Orthopedics, Artificial Joints Engineering and Technology Research Center of Jiangxi Province, The First Affiliated Hospital of Nanchang University, Nanchang University, 330006, Nanchang, Jiangxi, China.
| |
Collapse
|
28
|
Glushkova M, Yordanova I, Todorov T, Bojinova V, Koleva M, Dimova P, Tournev I, Angelova L, Todorova A, Mitev V. Three Novel NF1 Gene Mutations in a Cohort of Bulgarian Neurofibromatoses Patients. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418010040] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
29
|
Huang L, Ding Y, Qi L, Wu X, Li W, Huang G, Dai M, Zhang B. Repeated Multiple Neurofibromatosis Type 1 in the Right Lower Limb: A Case Report. World J Oncol 2017; 8:58-61. [PMID: 29147436 PMCID: PMC5649998 DOI: 10.14740/wjon1011w] [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] [Accepted: 04/18/2017] [Indexed: 11/26/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal-dominant genetic disease characterized by the presence of multiple neurofibromas. We encountered a unique case of NF1 that manifested as a recurrent soft tissue neurofibroma in the right lower limb that developed over a period of 16 years. The patient presented with a painless mass that was initially diagnosed as inflammatory changes via computed tomography and magnetic resonance imaging. However, the condition was subsequently diagnosed as an intraneural neurofibroma via pathological and immunohistochemical examination, which showed a focal to patchy lymphocytic chronic inflammatory infiltrate and several non-encapsulated masses with clear boundaries that were easily distinguishable from the adjacent neurofibroma. The mass relapsed three times over 3 years since it was discovered, for which the patient underwent comprehensive and complete local resection several times. Postoperative continuous follow-up confirmed that the patient recovered well. Early and complete surgical resection is an effective method for treating and preventing recurrent neurofibromas. However, because of the importance of pathologic examination in the diagnosis of such cases, this uncommon entity might be underreported in patients with NF1.
Collapse
Affiliation(s)
- Leitao Huang
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China.,These authors contributed equally to this work
| | - Yi Ding
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China.,These authors contributed equally to this work
| | - Lai Qi
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xia Wu
- Department of College of Pharmacy, Nanchang University, Nanchang 330006, China
| | - Wei Li
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gendong Huang
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Min Dai
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Bing Zhang
- Department of Orthopedics, Artificial Joints Engineering and Technology, Research Center of Jiangxi Province, Nanchang, China.,Department of Orthopedics, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
30
|
McConnell MJ, Moran JV, Abyzov A, Akbarian S, Bae T, Cortes-Ciriano I, Erwin JA, Fasching L, Flasch DA, Freed D, Ganz J, Jaffe AE, Kwan KY, Kwon M, Lodato MA, Mills RE, Paquola ACM, Rodin RE, Rosenbluh C, Sestan N, Sherman MA, Shin JH, Song S, Straub RE, Thorpe J, Weinberger DR, Urban AE, Zhou B, Gage FH, Lehner T, Senthil G, Walsh CA, Chess A, Courchesne E, Gleeson JG, Kidd JM, Park PJ, Pevsner J, Vaccarino FM. Intersection of diverse neuronal genomes and neuropsychiatric disease: The Brain Somatic Mosaicism Network. Science 2017; 356:356/6336/eaal1641. [PMID: 28450582 DOI: 10.1126/science.aal1641] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Neuropsychiatric disorders have a complex genetic architecture. Human genetic population-based studies have identified numerous heritable sequence and structural genomic variants associated with susceptibility to neuropsychiatric disease. However, these germline variants do not fully account for disease risk. During brain development, progenitor cells undergo billions of cell divisions to generate the ~80 billion neurons in the brain. The failure to accurately repair DNA damage arising during replication, transcription, and cellular metabolism amid this dramatic cellular expansion can lead to somatic mutations. Somatic mutations that alter subsets of neuronal transcriptomes and proteomes can, in turn, affect cell proliferation and survival and lead to neurodevelopmental disorders. The long life span of individual neurons and the direct relationship between neural circuits and behavior suggest that somatic mutations in small populations of neurons can significantly affect individual neurodevelopment. The Brain Somatic Mosaicism Network has been founded to study somatic mosaicism both in neurotypical human brains and in the context of complex neuropsychiatric disorders.
Collapse
|
31
|
Kehrer-Sawatzki H, Farschtschi S, Mautner VF, Cooper DN. The molecular pathogenesis of schwannomatosis, a paradigm for the co-involvement of multiple tumour suppressor genes in tumorigenesis. Hum Genet 2016; 136:129-148. [PMID: 27921248 PMCID: PMC5258795 DOI: 10.1007/s00439-016-1753-8] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Accepted: 11/27/2016] [Indexed: 12/20/2022]
Abstract
Schwannomatosis is characterized by the predisposition to develop multiple schwannomas and, less commonly, meningiomas. Despite the clinical overlap with neurofibromatosis type 2 (NF2), schwannomatosis is not caused by germline NF2 gene mutations. Instead, germline mutations of either the SMARCB1 or LZTR1 tumour suppressor genes have been identified in 86% of familial and 40% of sporadic schwannomatosis patients. In contrast to patients with rhabdoid tumours, which are due to complete loss-of-function SMARCB1 mutations, individuals with schwannomatosis harbour predominantly hypomorphic SMARCB1 mutations which give rise to the synthesis of mutant proteins with residual function that do not cause rhabdoid tumours. Although biallelic mutations of SMARCB1 or LZTR1 have been detected in the tumours of patients with schwannomatosis, the classical two-hit model of tumorigenesis is insufficient to account for schwannoma growth, since NF2 is also frequently inactivated in these tumours. Consequently, tumorigenesis in schwannomatosis must involve the mutation of at least two different tumour suppressor genes, an occurrence frequently mediated by loss of heterozygosity of large parts of chromosome 22q harbouring not only SMARCB1 and LZTR1 but also NF2. Thus, schwannomatosis is paradigmatic for a tumour predisposition syndrome caused by the concomitant mutational inactivation of two or more tumour suppressor genes. This review provides an overview of current models of tumorigenesis and mutational patterns underlying schwannomatosis that will ultimately help to explain the complex clinical presentation of this rare disease.
Collapse
Affiliation(s)
| | - Said Farschtschi
- Department of Neurology, University Hospital Hamburg Eppendorf, 20246, Hamburg, Germany
| | - Victor-Felix Mautner
- Department of Neurology, University Hospital Hamburg Eppendorf, 20246, Hamburg, Germany
| | - David N Cooper
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff, CF14 4XN, UK
| |
Collapse
|
32
|
Li K, Turner AN, Chen M, Brosius SN, Schoeb TR, Messiaen LM, Bedwell DM, Zinn KR, Anastasaki C, Gutmann DH, Korf BR, Kesterson RA. Mice with missense and nonsense NF1 mutations display divergent phenotypes compared with human neurofibromatosis type I. Dis Model Mech 2016; 9:759-67. [PMID: 27482814 PMCID: PMC4958313 DOI: 10.1242/dmm.025783] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2016] [Accepted: 05/25/2016] [Indexed: 12/19/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a common genetic disorder characterized by the occurrence of nerve sheath tumors and considerable clinical heterogeneity. Some translational studies have been limited by the lack of animal models available for assessing patient-specific mutations. In order to test therapeutic approaches that might restore function to the mutated gene or gene product, we developed mice harboring NF1 patient-specific mutations including a nonsense mutation (c.2041C>T; p.Arg681*) and a missense mutation (c.2542G>C; p.Gly848Arg). The latter is associated with the development of multiple plexiform neurofibromas along spinal nerve roots. We demonstrate that the human nonsense NF1(Arg681*) and missense NF1(Gly848Arg) mutations have different effects on neurofibromin expression in the mouse and each recapitulates unique aspects of the NF1 phenotype, depending upon the genetic context when assessed in the homozygous state or when paired with a conditional knockout allele. Whereas the missense Nf1(Gly848Arg) mutation fails to produce an overt phenotype in the mouse, animals homozygous for the nonsense Nf1(Arg681*) mutation are not viable. Mice with one Nf1(Arg681*) allele in combination with a conditional floxed Nf1 allele and the DhhCre transgene (Nf1(4F/Arg681*); DhhCre) display disorganized nonmyelinating axons and neurofibromas along the spinal column, which leads to compression of the spinal cord and paralysis. This model will be valuable for preclinical testing of novel nonsense suppression therapies using drugs to target in-frame point mutations that create premature termination codons in individuals with NF1.
Collapse
Affiliation(s)
- Kairong Li
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ashley N Turner
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Min Chen
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Stephanie N Brosius
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA Medical Scientist Training Program, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Trenton R Schoeb
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Ludwine M Messiaen
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - David M Bedwell
- Department of Biochemistry and Molecular Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Kurt R Zinn
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bruce R Korf
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Robert A Kesterson
- Department of Genetics, The University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
33
|
Yap P, Super L, Qin J, Burgess T, Prodanovic Z, Edwards C, Thomas R, Carpenter K, Tan TY. Congenital Retroperitoneal Teratoma in Neurofibromatosis Type 1. Pediatr Blood Cancer 2016; 63:706-8. [PMID: 26514327 DOI: 10.1002/pbc.25812] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Revised: 09/29/2015] [Accepted: 09/30/2015] [Indexed: 12/11/2022]
Abstract
Neurofibromatosis type 1 (NF1) is caused by mutations in the tumor suppressor gene NF1. The increased tumor risk in affected individuals is well established, caused by somatic biallelic inactivation of NF1 due to loss of heterozygosity. Pediatric teratoma has not been reported in individuals with NF1 previously. We report a case of congenital teratoma in an infant with a heterozygous maternally inherited pathogenic NF1 mutation (c.[1756_1759delACTA] and p.[Thr586Valfs*18]). We detected a "second hit" in the form of mosaic whole NF1 deletion in the tumor tissue using multiplex ligation-dependent probe amplification, as a proof to support the hypothesis of NF1 involvement in the pathogenesis of teratoma.
Collapse
Affiliation(s)
- Patrick Yap
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia
| | - Leanne Super
- Monash Children's Cancer Centre, Monash Medical Centre, Clayton, Victoria, Australia
| | - Jinyi Qin
- Department of Diagnostic Genomics, PathWest Laboratory Medicine, Western Australia, Australia
| | - Trent Burgess
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, Royal Children's Hospital, University of Melbourne, Melbourne, Australia
| | | | - Caitlin Edwards
- Department of Diagnostic Genomics, PathWest Laboratory Medicine, Western Australia, Australia
| | - Rosemary Thomas
- Monash Children's Cancer Centre, Monash Medical Centre, Clayton, Victoria, Australia
| | - Karen Carpenter
- Department of Diagnostic Genomics, PathWest Laboratory Medicine, Western Australia, Australia
| | - Tiong Yang Tan
- Victorian Clinical Genetics Services, Murdoch Children's Research Institute, Melbourne, Australia
- Department of Paediatrics, Royal Children's Hospital, University of Melbourne, Melbourne, Australia
| |
Collapse
|
34
|
Happle R. The categories of cutaneous mosaicism: A proposed classification. Am J Med Genet A 2015; 170A:452-459. [PMID: 26494396 DOI: 10.1002/ajmg.a.37439] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Accepted: 10/05/2015] [Indexed: 11/08/2022]
Abstract
Mosaic disorders can most easily be studied in the skin. This article presents a comprehensive overview of the different forms of cutaneous mosaicism. Major categories are genomic versus epigenetic mosaicism and nonsegmental versus segmental mosaicism. The class of nonsegmental mosaics includes single point mosaicism as exemplified by solitary benign or malignant skin tumors; disseminated mosaicism as noted in autosomal dominant tumor syndromes such as neurofibromatosis 1; and patchy mosaicism without midline separation as found in giant melanocytic nevus. The class of segmental mosaics includes segmental manifestation of lethal genes surviving by mosaicism as noted in Proteus syndrome; type 1 segmental mosaicism of autosomal dominant skin disorders reflecting heterozygosity for a postzygotic new mutation; type 2 segmental mosaicism of autosomal dominant skin disorders reflecting loss of heterozygosity that occurred at an early developmental stage in a heterozygous embryo; and isolated or superimposed segmental mosaicism of common polygenic skin disorders such as psoriasis or atopic dermatitis. A particular form of genomic mosaicism is didymosis (twin spotting). Revertant mosaicism is recognizable as one or more areas of healthy skin in patients with epidermolysis bullosa or other serious genodermatoses. The category of epigenetic mosaicism includes several X-linked, male lethal disorders such as incontinentia pigmenti, and the patterns of lyonization as noted in X-linked non-lethal disorders such as hypohidrotic ectodermal dysplasia of the Christ-Siemens-Touraine type. An interesting field of future research will be the concept of epigenetic autosomal mosaicism that may explain some unusual cases of autosomal transmission of linear hypo- or hypermelanosis.
Collapse
Affiliation(s)
- Rudolf Happle
- Department of Dermatology, Freiburg University Medical Center, Freiburg, Germany
| |
Collapse
|
35
|
|
36
|
Rodrigues LN, Correa GB, Diniz MG, Galvão CF, Gomes CC, Gomez RS. BRAF V600E and loss of heterozygosity assessment in benign oralneural tumours. J Oral Pathol Med 2015; 44:634-7. [DOI: 10.1111/jop.12337] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/18/2015] [Indexed: 02/04/2023]
Affiliation(s)
- Leonardo Nogueira Rodrigues
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - GefterThiago Batista Correa
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - Marina Gonçalves Diniz
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - Clarice Ferreira Galvão
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - Carolina Cavaliéri Gomes
- Department of Pathology; Biological Sciences Institute; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| | - Ricardo Santiago Gomez
- Department of Oral Surgery and Pathology; School of Dentistry; Universidade Federal de Minas Gerais; Belo Horizonte Brazil
| |
Collapse
|
37
|
Somatic mutations, genome mosaicism, cancer and aging. Curr Opin Genet Dev 2014; 26:141-9. [PMID: 25282114 DOI: 10.1016/j.gde.2014.04.002] [Citation(s) in RCA: 99] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2014] [Revised: 04/04/2014] [Accepted: 04/11/2014] [Indexed: 01/11/2023]
Abstract
Genomes are inherently unstable due to the need for DNA sequence variation in the germ line to fuel evolution through natural selection. In somatic tissues mutations accumulate during development and aging, generating genome mosaics. There is little information about the possible causal role of increased somatic mutation loads in late-life disease and aging, with the exception of cancer. Characterizing somatic mutations and their functional consequences in normal tissues remains a formidable challenge due to their low, individual abundance. Here, I will briefly review our current knowledge of somatic mutations in animals and humans in relation to aging, how they arise and lead to genome mosaicism, the technology to study somatic mutations and how they possibly could cause non-clonal disease.
Collapse
|
38
|
Giovannoni I, Callea F, Boldrini R, Inserra A, Cozza R, Francalanci P. Malignant pheochromocytoma in a 16-year-old patient with neurofibromatosis type 1. Pediatr Dev Pathol 2014; 17:126-9. [PMID: 24555864 DOI: 10.2350/13-10-1397-cr.1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Patients with neurofibromatosis type I (NF1) feature a high risk of developing benign and malignant tumors, mainly those with a neuroectodermal origin, the risk being about 4 times higher than in the general population. Pheochromocytoma (PHEO) is a sporadic tumor (1∶100,000) arising from the adrenal medulla. Pheochromocytoma is a rare condition when occurring in conjunction with NF1 and occurs in about 1% of patients, rarely in those of pediatric age. In this study we present a 16-year-old patient with NF1 and malignant PHEO. Loss of heterozygosity analysis in PHEOs shows a reduction to homozygosity, observed for both 17p and 17q markers. This case confirms the importance of surveillance for malignant neoplasias in NF1 patients during childhood and adolescence. On the other hand, since 30% of PHEOs had germline mutations and, more rarely, somatic mutations, patients with PHEO should be investigated for associated genetic syndromes.
Collapse
Affiliation(s)
- Isabella Giovannoni
- 1 Department of Pathology, Children's Hospital Bambino Gesù, IRCCS Rome, Italy
| | | | | | | | | | | |
Collapse
|
39
|
Brain somatic mutations: the dark matter of psychiatric genetics? Mol Psychiatry 2014; 19:156-8. [PMID: 24342990 DOI: 10.1038/mp.2013.168] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2013] [Revised: 10/10/2013] [Accepted: 10/21/2013] [Indexed: 02/02/2023]
Abstract
Although inherited DNA sequences have a well-demonstrated role in psychiatric disease risk, for even the most heritable mental disorders, monozygotic twins are discordant at a significant rate. The genetic variation associated with mental disorders has heretofore been based on the search for rare or common variation in blood cells. This search is based on the premise that every somatic cell shares an identical DNA sequence, so that variation found in lymphocytes should reflect variation present in brain cells. Evidence from the study of cancer cells, stem cells and now neurons demonstrate that this premise is false. Somatic mutation is common in human cells and has been implicated in a range of diseases beyond cancer. The exuberant proliferation of cortical precursors during fetal development provides a likely environment for somatic mutation in neuronal and glial lineages. Studies of rare neurodevelopmental disorders, such as hemimegencephaly, demonstrate somatic mutations in affected cortical cells that cannot be detected in unaffected parts of the brain or in peripheral cells. This perspective argues for the need to investigate somatic variation in the brain as an explanation of the discordance in monozygotic twins, a proximate cause of mental disorders in individuals with inherited risk, and a potential guide to novel treatment targets.
Collapse
|
40
|
Ishida M, Okabe H. Cutaneous squamous cell carcinoma in a patient with neurofibromatosis type 1: A case report. Oncol Lett 2013; 6:878-880. [PMID: 24137429 PMCID: PMC3796415 DOI: 10.3892/ol.2013.1490] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2013] [Accepted: 07/15/2013] [Indexed: 11/06/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant inherited disease that is characterized by the presence of multiple neurofibromas, café-au-lait spots and iris hamartomas. It is well established that the incidence of tumors in patients with NF1 is high compared with the normal population and that the majority of the tumors are non-epithelial neoplasms, including neurofibromas, malignant peripheral nerve sheath tumors, gliomas and leukemia. Studies have suggested that patients with NF1 also have a significantly higher risk of certain types of carcinomas. However, the occurrence of cutaneous squamous cell carcinoma (SCC) in a patient with NF1 is extremely rare. The present study describes the second documented case of a cutaneous SCC adjacent to a neurofibroma of the forehead with histopathological analyses in a patient with NF1. An 80-year-old female with NF1 presented with a rapidly growing skin tumor of the forehead. Histopathological study of the resected forehead tumor demonstrated that there were two tumorous lesions. One was an invasive SCC and the other was a neurofibroma. The lesions were adjacent, but no continuity was present. NF1 is caused by inactivating mutations in the NF1 gene and loss of heterozygosity of this gene has been reported in neurofibromas, malignant peripheral nerve sheath tumors, gliomas and pheochromocytomas in patients with NF1. However, the genetic mechanism of carcinoma development in patients with NF1 is not well understood. Studies have suggested the role of the NF1 and/or the BRCA gene in the occurrence of breast cancer. Additional studies are required to elucidate these mechanisms.
Collapse
Affiliation(s)
- Mitsuaki Ishida
- Department of Clinical Laboratory Medicine and Division of Diagnostic Pathology, Shiga University of Medical Science, Otsu, Shiga, Japan
| | | |
Collapse
|
41
|
Poduri A, Evrony GD, Cai X, Walsh CA. Somatic mutation, genomic variation, and neurological disease. Science 2013; 341:1237758. [PMID: 23828942 DOI: 10.1126/science.1237758] [Citation(s) in RCA: 421] [Impact Index Per Article: 35.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Genetic mutations causing human disease are conventionally thought to be inherited through the germ line from one's parents and present in all somatic (body) cells, except for most cancer mutations, which arise somatically. Increasingly, somatic mutations are being identified in diseases other than cancer, including neurodevelopmental diseases. Somatic mutations can arise during the course of prenatal brain development and cause neurological disease-even when present at low levels of mosaicism, for example-resulting in brain malformations associated with epilepsy and intellectual disability. Novel, highly sensitive technologies will allow more accurate evaluation of somatic mutations in neurodevelopmental disorders and during normal brain development.
Collapse
Affiliation(s)
- Annapurna Poduri
- Department of Neurology, Boston Children's Hospital, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
42
|
Spaepen M, Neven E, Sagaert X, De Hertogh G, Beert E, Wimmer K, Matthijs G, Legius E, Brems H. EPCAM germline and somatic rearrangements in Lynch syndrome: identification of a novel 3'EPCAM deletion. Genes Chromosomes Cancer 2013; 52:845-54. [PMID: 23801599 DOI: 10.1002/gcc.22080] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Revised: 05/15/2013] [Accepted: 05/16/2013] [Indexed: 11/07/2022] Open
Abstract
3'EPCAM (Epithelial Cell Adhesion Molecule) genomic rearrangements can be a cause of mismatch repair deficiency in rare Lynch syndrome families. 3'EPCAM deletions include the polyadenylation signal and might result in promoter hypermethylation of the centromeric MSH2 gene in cis. A somatic rearrangement in trans affecting MSH2 is responsible for the final mismatch repair deficiency in the corresponding tumors but the mechanisms are not well documented. In this report two germline 3'EPCAM deletions are described together with the corresponding somatic mutations in the patient's colorectal tumors. Mutation and breakpoint analysis resulted in the identification of one novel (c.556-531_*872del) and one known EPCAM deletion (c.859-689_*14697del). Both deletions resulted from Alu mediated homologous recombination causing aberrant EPCAM-MSH2 fusion transcripts. The colorectal tumors of the deletion carriers were MSI-high. Strong hypermethylation of the MSH2 promoter was measured. Analysis of somatic genomic rearrangements showed a 4 Mb deletion including the EPCAM, MSH2 and MSH6 genes in one tumor and copy neutral loss of heterozygosity in the EPCAM-MSH2 region in the other tumor. This indicates that hemi- and homozygous hypermethylation of the MSH2 promoter and hence complete silencing of MSH2 expression was responsible for the mismatch repair deficiency in both colorectal tumors.
Collapse
Affiliation(s)
- Marijke Spaepen
- Department of Human Genetics, University Hospital Leuven, 3000 Leuven, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sant'Anna JR, Yajima JPRS, Rosada LJ, Franco CCS, Prioli AJ, Della-Rosa VA, Mathias PCF, Castro-Prado MAA. Metformin's performance in in vitro and in vivo genetic toxicology studies. Exp Biol Med (Maywood) 2013; 238:803-10. [PMID: 23788173 DOI: 10.1177/1535370213480744] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Metformin is a hypoglycemiant drug prescribed for the treatment and control of the type 2 diabetes mellitus. Recently, the potential efficacy of this antidiabetic drug as an anticancer agent has been demonstrated in various mammalian cancer cells. This report evaluates the mutagenic as well as the recombinogenic potentials of the metformin drug in therapeutically relevant plasma concentrations (12.5 µM, 25.0 µM or 50.0 µM). Since the loss of heterozygosity is a process associated with carcinogenesis, the recombinogenic potential of such a drug was evaluated by the homozygotization assay using a heterozygous diploid strain of Aspergillus nidulans. The homozigotization indices (HI) for the genetic markers from the metformin-treated diploids were not statistically different from the negative control (non-treated diploids). For the first time, this indicated a lack of recombinogenic activity of the antidiabetic drug. The mutagenic potential of the metformin drug was evaluated by the chromosome aberrations and the micronuclei tests in human lymphocytes cultures. The metformin drug did not show any significant increase either in the numerical or in the structural chromosome aberrations and did not affect significantly the mitotic index when compared to the negative control. In the in vitro micronucleus test, the drug did not increase the number of micronuclei or nuclear buds when compared with the negative control. The data in this study suggest that the metformin drug is not a secondary cancer inducer, since it has neither showed recombinogenic nor mutagenic activities when used in pharmacological concentrations.
Collapse
Affiliation(s)
- Juliane R Sant'Anna
- Laboratório de Genética de Microorganismos e Mutagênese, Departamento de Biotecnologia, Genética e Biologia Celular, Universidade Estadual de Maringá, 87020-900, Maringá, PR, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Terribas E, Garcia-Linares C, Lázaro C, Serra E. Probe-based quantitative PCR assay for detecting constitutional and somatic deletions in the NF1 gene: application to genetic testing and tumor analysis. Clin Chem 2013; 59:928-37. [PMID: 23386700 DOI: 10.1373/clinchem.2012.194217] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND About 5% of patients with neurofibromatosis type 1 (NF1) bear constitutional microdeletions that encompass NF1 (neurofibromin 1) and neighboring genes. These patients are characterized by the development of a high number of dermal neurofibromas (dNFs), mental retardation, and an increased risk of developing a malignant peripheral nerve sheath tumor (MPNST). Additionally, 10% of somatic second hits identified in dNFs are caused by deletions involving the NF1 gene. To detect constitutional and somatic deletions, we developed a probe-based quantitative PCR (qPCR) assay for interrogating the copy number status of 11 loci distributed along a 2.8-Mb region around the NF1 gene. METHODS We developed the qPCR assay with Universal ProbeLibrary technology (Roche) and designed a Microsoft Excel spreadsheet to analyze qPCR data for copy number calculations. The assay fulfilled the essential aspects of the MIQE (minimum information for publication of quantitative real-time PCR experiments) guidelines and used the qBase relative quantification framework for calculations. RESULTS The assay was validated with a set of DNA samples with known constitutional or somatic NF1 deletions. The assay showed high diagnostic sensitivity and specificity and distinguished between Type-1, Type-2, and atypical constitutional microdeletions in 14 different samples. It also identified 16 different somatic deletions in dNFs. These results were confirmed by multiplex ligation-dependent probe amplification. CONCLUSIONS The qPCR assay provides a methodology for detecting constitutional NF1 microdeletions that could be incorporated as an additional technique in a genetic-testing setting. It also permits the identification of somatic NF1 deletions in tissues with a high percentage of cells bearing 2 copies of the NF1 gene.
Collapse
Affiliation(s)
- Ernest Terribas
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
| | | | | | | |
Collapse
|
45
|
Thomas L, Mautner VF, Cooper DN, Upadhyaya M. Molecular heterogeneity in malignant peripheral nerve sheath tumors associated with neurofibromatosis type 1. Hum Genomics 2012; 6:18. [PMID: 23244685 PMCID: PMC3500234 DOI: 10.1186/1479-7364-6-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Accepted: 07/08/2012] [Indexed: 12/21/2022] Open
Abstract
Neurofibromatosis type-1 (NF1), resulting from NF1 gene loss of function, is characterized by an increased risk of developing benign and malignant peripheral nerve sheath tumors (MPNSTs). Whereas the cellular heterogeneity of NF1-associated tumors has been well studied, the molecular heterogeneity of MPNSTs is still poorly understood. Mutational heterogeneity within these malignant tumors greatly complicates the study of the underlying mechanisms of tumorigenesis. We have explored this molecular heterogeneity by performing loss of heterozygosity (LOH) analysis of the NF1, TP53, RB1, PTEN, and CDKN2A genes on sections of 10 MPNSTs derived from 10 unrelated NF1 patients. LOH data for the TP53 gene was found to correlate with the results of p53 immunohistochemical analysis in the same tumor sections. Further, approximately 70% of MPNSTs were found to display intra-tumoral molecular heterogeneity as evidenced by differences in the level of LOH between different sections of the same tumor samples. This study constitutes the first systematic analysis of molecular heterogeneity within MPNSTs derived from NF1 patients. Appreciation of the existence of molecular heterogeneity in NF1-associated tumors is important not only for optimizing somatic mutation detection, but also for understanding the mechanisms of NF1 tumorigenesis, a prerequisite for the development of specifically targeted cancer therapeutics.
Collapse
Affiliation(s)
- Laura Thomas
- Institute of Medical Genetics, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| | | | | | | |
Collapse
|
46
|
Garcia-Linares C, Mercadé J, Gel B, Biayna J, Terribas E, Lázaro C, Serra E. Applying microsatellite multiplex PCR analysis (MMPA) for determining allele copy-number status and percentage of normal cells within tumors. PLoS One 2012; 7:e42682. [PMID: 22916147 PMCID: PMC3419736 DOI: 10.1371/journal.pone.0042682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2011] [Accepted: 07/11/2012] [Indexed: 11/18/2022] Open
Abstract
The study of somatic genetic alterations in tumors contributes to the understanding and management of cancer. Genetic alterations, such us copy number or copy neutral changes, generate allelic imbalances (AIs) that can be determined using polymorphic markers. Here we report the development of a simple set of calculations for analyzing microsatellite multiplex PCR data from control-tumor pairs that allows us to obtain accurate information not only regarding the AI status of tumors, but also the percentage of tumor-infiltrating normal cells, the locus copy-number status and the mechanism involved in AI. We validated this new approach by re-analyzing a set of Neurofibromatosis type 1-associated dermal neurofibromas and comparing newly generated data with results obtained for the same tumors in a previous study using MLPA, Paralog Ratio Analysis and SNP-array techniques.Microsatellite multiplex PCR analysis (MMPA) should be particularly useful for analyzing specific regions of the genome containing tumor suppressor genes and also for determining the percentage of infiltrating normal cells within tumors allowing them to be sorted before they are analyzed by more expensive techniques.
Collapse
Affiliation(s)
- Carles Garcia-Linares
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
| | - Jaume Mercadé
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
| | - Bernat Gel
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
| | - Josep Biayna
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
| | - Ernest Terribas
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
| | - Conxi Lázaro
- Programa de Diagnòstic Molecular de Càncer Hereditari, Laboratori de Recerca Translacional, Institut Català d'Oncologia (ICO) – IDIBELL, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Eduard Serra
- Institute of Predictive and Personalized Medicine of Cancer (IMPPC), Badalona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
47
|
Patel AV, Eaves D, Jessen WJ, Rizvi TA, Ecsedy JA, Qian MG, Aronow BJ, Perentesis JP, Serra E, Cripe TP, Miller SJ, Ratner N. Ras-driven transcriptome analysis identifies aurora kinase A as a potential malignant peripheral nerve sheath tumor therapeutic target. Clin Cancer Res 2012; 18:5020-30. [PMID: 22811580 DOI: 10.1158/1078-0432.ccr-12-1072] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
PURPOSE Patients with neurofibromatosis type 1 (NF1) develop malignant peripheral nerve sheath tumors (MPNST), which are often inoperable and do not respond well to current chemotherapies or radiation. The goal of this study was to use comprehensive gene expression analysis to identify novel therapeutic targets. EXPERIMENTAL DESIGN Nerve Schwann cells and/or their precursors are the tumorigenic cell types in MPNST because of the loss of the NF1 gene, which encodes the RasGAP protein neurofibromin. Therefore, we created a transgenic mouse model, CNP-HRas12V, expressing constitutively active HRas in Schwann cells and defined a Ras-induced gene expression signature to drive a Bayesian factor regression model analysis of differentially expressed genes in mouse and human neurofibromas and MPNSTs. We tested functional significance of Aurora kinase overexpression in MPNST in vitro and in vivo using Aurora kinase short hairpin RNAs (shRNA) and compounds that inhibit Aurora kinase. RESULTS We identified 2,000 genes with probability of linkage to nerve Ras signaling of which 339 were significantly differentially expressed in mouse and human NF1-related tumor samples relative to normal nerves, including Aurora kinase A (AURKA). AURKA was dramatically overexpressed and genomically amplified in MPNSTs but not neurofibromas. Aurora kinase shRNAs and Aurora kinase inhibitors blocked MPNST cell growth in vitro. Furthermore, an AURKA selective inhibitor, MLN8237, stabilized tumor volume and significantly increased survival of mice with MPNST xenografts. CONCLUSION Integrative cross-species transcriptome analyses combined with preclinical testing has provided an effective method for identifying candidates for molecular-targeted therapeutics. Blocking Aurora kinases may be a viable treatment platform for MPNST.
Collapse
Affiliation(s)
- Ami V Patel
- Divisions of Experimental Hematology and Cancer Biology, Oncology, and Biomedical Informatics, Cincinnati Children's Hospital Medical Center, Cincinnati, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Laycock-van Spyk S, Thomas N, Cooper DN, Upadhyaya M. Neurofibromatosis type 1-associated tumours: their somatic mutational spectrum and pathogenesis. Hum Genomics 2012; 5:623-90. [PMID: 22155606 PMCID: PMC3525246 DOI: 10.1186/1479-7364-5-6-623] [Citation(s) in RCA: 101] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Somatic gene mutations constitute key events in the malignant transformation of human cells. Somatic mutation can either actively speed up the growth of tumour cells or relax the growth constraints normally imposed upon them, thereby conferring a selective (proliferative) advantage at the cellular level. Neurofibromatosis type-1 (NF1) affects 1/3,000-4,000 individuals worldwide and is caused by the inactivation of the NF1 tumour suppressor gene, which encodes the protein neurofibromin. Consistent with Knudson's two-hit hypothesis, NF1 patients harbouring a heterozygous germline NF1 mutation develop neurofibromas upon somatic mutation of the second, wild-type, NF1 allele. While the identification of somatic mutations in NF1 patients has always been problematic on account of the extensive cellular heterogeneity manifested by neurofibromas, the classification of NF1 somatic mutations is a prerequisite for understanding the complex molecular mechanisms underlying NF1 tumorigenesis. Here, the known somatic mutational spectrum for the NF1 gene in a range of NF1-associated neoplasms --including peripheral nerve sheath tumours (neurofibromas), malignant peripheral nerve sheath tumours, gastrointestinal stromal tumours, gastric carcinoid, juvenile myelomonocytic leukaemia, glomus tumours, astrocytomas and phaeochromocytomas -- have been collated and analysed.
Collapse
|
49
|
Stewart DR, Pemov A, Van Loo P, Beert E, Brems H, Sciot R, Claes K, Pak E, Dutra A, Lee CCR, Legius E. Mitotic recombination of chromosome arm 17q as a cause of loss of heterozygosity of NF1 in neurofibromatosis type 1-associated glomus tumors. Genes Chromosomes Cancer 2012; 51:429-37. [PMID: 22250039 PMCID: PMC3295917 DOI: 10.1002/gcc.21928] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2011] [Accepted: 12/12/2011] [Indexed: 12/16/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a common, autosomal dominant, tumor-predisposition syndrome that arises secondary to mutations in NF1. Glomus tumors are painful benign tumors that originate from the glomus body in the fingers and toes due to biallelic inactivation of NF1. We karyotyped cultures from four previously reported and one new glomus tumor and hybridized tumor (and matching germline) DNA on Illumina HumanOmni1-Quad SNP arrays (≈ 1 × 10(6) SNPs). Two tumors displayed evidence of copy-neutral loss of heterozygosity of chromosome arm 17q not observed in the germline sample, consistent with a mitotic recombination event. One of these two tumors, NF1-G12, featured extreme polyploidy (near-tetraploidy, near-hexaploidy, or near-septaploidy) across all chromosomes. In the remaining four tumors, there were few cytogenetic abnormalities observed, and copy-number analysis was consistent with diploidy in all chromosomes. This is the first study of glomus tumors cytogenetics, to our knowledge, and the first to report biallelic inactivation of NF1 secondary to mitotic recombination of chromosome arm 17q in multiple NF1-associated glomus tumors. We have observed mitotic recombination in 22% of molecularly characterized NF1-associated glomus tumors, suggesting that it is a not uncommon mechanism in the reduction to homozygosity of the NF1 germline mutation in these tumors. In tumor NF1-G12, we hypothesize that mitotic recombination also "unmasked" (reduced to homozygosity) a hypomorphic germline allele in a gene on chromosome arm 17q associated with chromosomal instability, resulting in the extreme polyploidy.
Collapse
Affiliation(s)
- Douglas R Stewart
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, Rockville, MD 20852, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Microarray-based copy number analysis of neurofibromatosis type-1 (NF1)-associated malignant peripheral nerve sheath tumors reveals a role for Rho-GTPase pathway genes in NF1 tumorigenesis. Hum Mutat 2012; 33:763-76. [DOI: 10.1002/humu.22044] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Accepted: 01/18/2012] [Indexed: 01/22/2023]
|