1
|
Lozano LP, Jensen R, Jennisch M, Pandala NG, Jamshidi F, Boldt HC, Tucker BA, Binkley EM. Genetics and current research models of Mendelian tumor predisposition syndromes with ocular involvement. Prog Retin Eye Res 2025; 106:101359. [PMID: 40274012 DOI: 10.1016/j.preteyeres.2025.101359] [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: 01/09/2025] [Revised: 04/17/2025] [Accepted: 04/18/2025] [Indexed: 04/26/2025]
Abstract
In this review, we aim to provide a survey of hereditable tumor predisposition syndromes with a Mendelian inheritance pattern and ocular involvement. We focus our discussion on von Hippel-Lindau disease, neurofibromatosis type 1, NF2-related schwannomatosis, tuberous sclerosis complex, retinoblastoma, and the BAP1 tumor predisposition syndrome. For each of the six diseases, we discuss the clinical presentation and the molecular pathophysiology. We emphasize the genetics, current research models, and therapeutic developments. After reading each disease section, readers should possess an understanding of the clinical presentation, genetic causes and inheritance patterns, and current state of research in disease modeling and treatment.
Collapse
Affiliation(s)
- Lola P Lozano
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Renato Jensen
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Madeleine Jennisch
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Narendra G Pandala
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA.
| | - Farzad Jamshidi
- Department of Ophthalmology, University of Pittsburgh/UPMC, Pittsburgh, PA, 15213, USA.
| | - H Culver Boldt
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Budd A Tucker
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| | - Elaine M Binkley
- Institute for Vision Research, The University of Iowa, Iowa City, IA, 52242, USA; Department of Ophthalmology and Visual Sciences, The University of Iowa Carver College of Medicine, Iowa City, IA, 52242, USA.
| |
Collapse
|
2
|
Oliver TRW, Lawson ARJ, Lee-Six H, Tollit A, Jung H, Hooks Y, Sanghvi R, Young MD, Butler TM, Nicola PA, Treger TD, Lensing SV, Burke GAA, Aquilina K, Löbel U, Cortes-Ciriano I, Hargrave D, Jorgensen M, Jessop FA, Coorens THH, Flanagan AM, Allinson K, Martincorena I, Jacques TS, Behjati S. Cancer-independent somatic mutation of the wild-type NF1 allele in normal tissues in neurofibromatosis type 1. Nat Genet 2025; 57:515-521. [PMID: 40000831 PMCID: PMC11906363 DOI: 10.1038/s41588-025-02097-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/22/2025] [Indexed: 02/27/2025]
Abstract
Cancer predisposition syndromes mediated by recessive cancer genes generate tumors via somatic variants (second hits) in the unaffected allele. Second hits may or may not be sufficient for neoplastic transformation. Here we performed whole-genome and whole-exome sequencing on 479 tissue biopsies from a child with neurofibromatosis type 1, a multisystem cancer-predisposing syndrome mediated by constitutive monoallelic NF1 inactivation. We identified multiple independent NF1 driver variants in histologically normal tissues, but not in 610 biopsies from two nonpredisposed children. We corroborated this finding using targeted duplex sequencing, including a further nine adults with the same syndrome. Overall, truncating NF1 mutations were under positive selection in normal tissues from individuals with neurofibromatosis type 1. We demonstrate that normal tissues in neurofibromatosis type 1 commonly harbor second hits in NF1, the extent and pattern of which may underpin the syndrome's cancer phenotype.
Collapse
Affiliation(s)
- Thomas R W Oliver
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Henry Lee-Six
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Anna Tollit
- Research Department of Pathology, University College London, London, UK
| | | | | | | | | | | | | | - Taryn D Treger
- Wellcome Sanger Institute, Hinxton, UK
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Paediatrics, University of Cambridge, Cambridge, UK
| | | | - G A Amos Burke
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, UK
| | - Kristian Aquilina
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Ulrike Löbel
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Isidro Cortes-Ciriano
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Cambridge, UK
| | - Darren Hargrave
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Mette Jorgensen
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Flora A Jessop
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | | | - Adrienne M Flanagan
- Research Department of Pathology, University College London, London, UK
- Department of Histopathology, Royal National Orthopaedic Hospital NHS Trust, Middlesex, UK
| | - Kieren Allinson
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
| | | | - Thomas S Jacques
- Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
- UCL Great Ormond Street Institute of Child Health, London, UK.
| | - Sam Behjati
- Wellcome Sanger Institute, Hinxton, UK.
- Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK.
- Department of Paediatrics, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Senanayake J, Mattingly RR, Sundararaghavan HG. Electrical stimulation of Schwann cells on electrospun hyaluronic acid carbon nanotube fibers. PLoS One 2024; 19:e0308207. [PMID: 39110684 PMCID: PMC11305570 DOI: 10.1371/journal.pone.0308207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 07/18/2024] [Indexed: 08/10/2024] Open
Abstract
Neurofibromatosis Type 1 (NF1) is a complex genetic disorder characterized by the development of benign neurofibromas, which can cause significant morbidity in affected individuals. While the molecular mechanisms underlying NF1 pathogenesis have been extensively studied, the development of effective therapeutic strategies remains a challenge. This paper presents the development and validation of a novel biomaterial testing model to enhance our understanding of NF1 pathophysiology, disease mechanisms and evaluate potential therapeutic interventions. Our long-term goal is to develop an invitro model of NF1 to evaluate drug targets. We have developed an in vitro system to test the cellular behavior of NF1 patient derived cells on electroconductive aligned nanofibrous biomaterials with electrical stimulatory cues. We hypothesized that cells cultured on electroconductive biomaterial will undergo morphological changes and variations in cell proliferation that could be further enhanced with the combination of exogenous electrical stimulation (ES). In this study, we developed electrospun Hyaluronic Acid-Carbon Nanotube (HA-CNT) nanofiber scaffolds to mimic the axon's topographical and bioelectrical cues that influence neurofibroma growth and development. The cellular behavior was qualitatively and quantitively analyzed through immunofluorescent stains, Alamar blue assays and ELISA assays. Schwann cells from NF1 patients appear to have lost their ability to respond to electrical stimulation in the development and regeneration range, which was seen through changes in morphology, proliferation and NGF release. Without stimulation, the conductive material enhances NF1 SC behavior. Wild-type SC respond to electrical stimulation with increased cell proliferation and NGF release. Using this system, we can better understand the interaction between axons and SC that lead to tumor formation, homeostasis and regeneration.
Collapse
Affiliation(s)
- Judy Senanayake
- Department of Biomedical Engineering, Wayne State University, Detroit, MI, United States of America
| | - Raymond R. Mattingly
- Department of Pharmacology and Toxicology, East Carolina University, Greenville, NC, United States of America
| | | |
Collapse
|
4
|
Ruiz-García C, Lassaletta L, López-Larrubia P, Varela-Nieto I, Murillo-Cuesta S. Tumors of the nervous system and hearing loss: Beyond vestibular schwannomas. Hear Res 2024; 447:109012. [PMID: 38703433 DOI: 10.1016/j.heares.2024.109012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 05/06/2024]
Abstract
Hearing loss is a common side effect of many tumor treatments. However, hearing loss can also occur as a direct result of certain tumors of the nervous system, the most common of which are the vestibular schwannomas (VS). These tumors arise from Schwann cells of the vestibulocochlear nerve and their main cause is the loss of function of NF2, with 95 % of cases being sporadic and 5 % being part of the rare neurofibromatosis type 2 (NF2)-related Schwannomatosis. Genetic variations in NF2 do not fully explain the clinical heterogeneity of VS, and interactions between Schwann cells and their microenvironment appear to be critical for tumor development. Preclinical in vitro and in vivo models of VS are needed to develop prognostic biomarkers and targeted therapies. In addition to VS, other tumors can affect hearing. Meningiomas and other masses in the cerebellopontine angle can compress the vestibulocochlear nerve due to their anatomic proximity. Gliomas can disrupt several neurological functions, including hearing; in fact, glioblastoma multiforme, the most aggressive subtype, may exhibit early symptoms of auditory alterations. Besides, treatments for high-grade tumors, including chemotherapy or radiotherapy, as well as incomplete resections, can induce long-term auditory dysfunction. Because hearing loss can have an irreversible and dramatic impact on quality of life, it should be considered in the clinical management plan of patients with tumors, and monitored throughout the course of the disease.
Collapse
Affiliation(s)
- Carmen Ruiz-García
- Department of Otorhinolaryngology, La Paz University Hospital. Paseo La Castellana 261, Madrid 28046, Spain; Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Neuropathology of Hearing and Myelinopathies, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain; PhD Program in Medicine and Surgery, Autonomous University of Madrid, Madrid, Spain
| | - Luis Lassaletta
- Department of Otorhinolaryngology, La Paz University Hospital. Paseo La Castellana 261, Madrid 28046, Spain; Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Monforte de Lemos 9-11, Madrid 28029, Spain
| | - Pilar López-Larrubia
- Biomedical Magnetic Resonance, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain
| | - Isabel Varela-Nieto
- Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Neuropathology of Hearing and Myelinopathies, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Monforte de Lemos 9-11, Madrid 28029, Spain.
| | - Silvia Murillo-Cuesta
- Research in Otoneurosurgery. Hospital La Paz Institute for Health Research - IdiPAZ (La Paz University Hospital - Universidad Autónoma de Madrid), Paseo La Castellana 261, Madrid 28046, Spain; Neuropathology of Hearing and Myelinopathies, Instituto de Investigaciones Biomédicas Sols-Morreale, CSIC-UAM. Arturo Duperier 4, Madrid 28029, Spain; Biomedical Research Networking Centre On Rare Diseases (CIBERER), Institute of Health Carlos III, Monforte de Lemos 9-11, Madrid 28029, Spain.
| |
Collapse
|
5
|
Atsoniou K, Giannopoulou E, Georganta EM, Skoulakis EMC. Drosophila Contributions towards Understanding Neurofibromatosis 1. Cells 2024; 13:721. [PMID: 38667335 PMCID: PMC11048932 DOI: 10.3390/cells13080721] [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: 03/15/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 04/28/2024] Open
Abstract
Neurofibromatosis 1 (NF1) is a multisymptomatic disorder with highly variable presentations, which include short stature, susceptibility to formation of the characteristic benign tumors known as neurofibromas, intense freckling and skin discoloration, and cognitive deficits, which characterize most children with the condition. Attention deficits and Autism Spectrum manifestations augment the compromised learning presented by most patients, leading to behavioral problems and school failure, while fragmented sleep contributes to chronic fatigue and poor quality of life. Neurofibromin (Nf1) is present ubiquitously during human development and postnatally in most neuronal, oligodendrocyte, and Schwann cells. Evidence largely from animal models including Drosophila suggests that the symptomatic variability may reflect distinct cell-type-specific functions of the protein, which emerge upon its loss, or mutations affecting the different functional domains of the protein. This review summarizes the contributions of Drosophila in modeling multiple NF1 manifestations, addressing hypotheses regarding the cell-type-specific functions of the protein and exploring the molecular pathways affected upon loss of the highly conserved fly homolog dNf1. Collectively, work in this model not only has efficiently and expediently modelled multiple aspects of the condition and increased understanding of its behavioral manifestations, but also has led to pharmaceutical strategies towards their amelioration.
Collapse
Affiliation(s)
- Kalliopi Atsoniou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
- Laboratory of Experimental Physiology, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Eleni Giannopoulou
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
| | - Eirini-Maria Georganta
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
| | - Efthimios M. C. Skoulakis
- Institute for Fundamental Biomedical Research, Biomedical Sciences Research Center “Alexander Fleming”, 16672 Athens, Greece; (K.A.); (E.G.)
| |
Collapse
|
6
|
McLean DT, Meudt JJ, Lopez Rivera LD, Schomberg DT, Pavelec DM, Duellman TT, Buehler DG, Schwartz PB, Graham M, Lee LM, Graff KD, Reichert JL, Bon-Durant SS, Konsitzke CM, Ronnekleiv-Kelly SM, Shanmuganayagam D, Rubinstein CD. Single-cell RNA sequencing of neurofibromas reveals a tumor microenvironment favorable for neural regeneration and immune suppression in a neurofibromatosis type 1 porcine model. Front Oncol 2023; 13:1253659. [PMID: 37817770 PMCID: PMC10561395 DOI: 10.3389/fonc.2023.1253659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Accepted: 09/11/2023] [Indexed: 10/12/2023] Open
Abstract
Neurofibromatosis Type 1 (NF1) is one of the most common genetically inherited disorders that affects 1 in 3000 children annually. Clinical manifestations vary widely but nearly always include the development of cutaneous, plexiform and diffuse neurofibromas that are managed over many years. Recent single-cell transcriptomics profiling efforts of neurofibromas have begun to reveal cell signaling processes. However, the cell signaling networks in mature, non-cutaneous neurofibromas remain unexplored. Here, we present insights into the cellular composition and signaling within mature neurofibromas, contrasting with normal adjacent tissue, in a porcine model of NF1 using single-cell RNA sequencing (scRNA-seq) analysis and histopathological characterization. These neurofibromas exhibited classic diffuse-type histologic morphology and expected patterns of S100, SOX10, GFAP, and CD34 immunohistochemistry. The porcine mature neurofibromas closely resemble human neurofibromas histologically and contain all known cellular components of their human counterparts. The scRNA-seq confirmed the presence of all expected cell types within these neurofibromas and identified novel populations of fibroblasts and immune cells, which may contribute to the tumor microenvironment by suppressing inflammation, promoting M2 macrophage polarization, increasing fibrosis, and driving the proliferation of Schwann cells. Notably, we identified tumor-associated IDO1 +/CD274+ (PD-L1) + dendritic cells, which represent the first such observation in any NF1 animal model and suggest the role of the upregulation of immune checkpoints in mature neurofibromas. Finally, we observed that cell types in the tumor microenvironment are poised to promote immune evasion, extracellular matrix reconstruction, and nerve regeneration.
Collapse
Affiliation(s)
- Dalton T. McLean
- Biotechnology Center, University of Wisconsin–Madison, Madison, WI, United States
- Molecular & Environmental Toxicology Program, University of Wisconsin–Madison, Madison, WI, United States
| | - Jennifer J. Meudt
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Loren D. Lopez Rivera
- Molecular & Environmental Toxicology Program, University of Wisconsin–Madison, Madison, WI, United States
| | - Dominic T. Schomberg
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Derek M. Pavelec
- Biotechnology Center, University of Wisconsin–Madison, Madison, WI, United States
| | - Tyler T. Duellman
- Biotechnology Center, University of Wisconsin–Madison, Madison, WI, United States
| | - Darya G. Buehler
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Patrick B. Schwartz
- Molecular & Environmental Toxicology Program, University of Wisconsin–Madison, Madison, WI, United States
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Melissa Graham
- Research Animal Resources and Compliance (RARC), Office of the Vice Chancellor for Research and Graduate Education, University of Wisconsin–Madison, Madison, WI, United States
| | - Laura M. Lee
- Research Animal Resources and Compliance (RARC), Office of the Vice Chancellor for Research and Graduate Education, University of Wisconsin–Madison, Madison, WI, United States
| | - Keri D. Graff
- Swine Research and Teaching Center, Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Jamie L. Reichert
- Swine Research and Teaching Center, Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI, United States
| | - Sandra S. Bon-Durant
- Biotechnology Center, University of Wisconsin–Madison, Madison, WI, United States
| | - Charles M. Konsitzke
- Biotechnology Center, University of Wisconsin–Madison, Madison, WI, United States
| | - Sean M. Ronnekleiv-Kelly
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
| | - Dhanansayan Shanmuganayagam
- Molecular & Environmental Toxicology Program, University of Wisconsin–Madison, Madison, WI, United States
- Biomedical & Genomic Research Group, Department of Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, WI, United States
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, Madison, WI, United States
- Center for Biomedical Swine Research and Innovation, University of Wisconsin–Madison, Madison, WI, United States
| | - C. Dustin Rubinstein
- Biotechnology Center, University of Wisconsin–Madison, Madison, WI, United States
| |
Collapse
|
7
|
Lalancette E, Charlebois-Poirier AR, Agbogba K, Knoth IS, Jones EJH, Mason L, Perreault S, Lippé S. Steady-state visual evoked potentials in children with neurofibromatosis type 1: associations with behavioral rating scales and impact of psychostimulant medication. J Neurodev Disord 2022; 14:42. [PMID: 35869419 PMCID: PMC9306184 DOI: 10.1186/s11689-022-09452-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 07/11/2022] [Indexed: 11/20/2022] Open
Abstract
Background Neurofibromatosis type 1 (NF1) is a genetic disorder often associated with cognitive dysfunctions, including a high occurrence of deficits in visuoperceptual skills. The neural underpinnings of these visuoperceptual deficits are not fully understood. We used steady-state visual evoked potentials (SSVEPs) to investigate possible alterations in the synchronization of neural activity in the occipital cortex of children with NF1. Methods SSVEPs were measured using electroencephalography and compared between children with NF1 (n = 28) and neurotypical controls (n = 28) aged between 4 and 13 years old. SSVEPs were recorded during visual stimulation with coloured icons flickering at three different frequencies (6 Hz, 10 Hz, and 15 Hz) and analyzed in terms of signal-to-noise ratios. A mixed design ANCOVA was performed to compare SSVEP responses between groups at the three stimulation frequencies. Pearson’s correlations with levels of intellectual functioning as well as with symptoms of ADHD, ASD and emotional/behavioral problems were performed. The impact of psychostimulant medication on the SSVEP responses was analyzed in a subset of the NF1 group (n = 8) with paired t-tests. Results We observed reduced signal-to-noise ratios of the SSVEP responses in children with NF1. The SSVEP responses were negatively correlated with symptoms of inattention and with symptoms of emotional/behavioral problems in the NF1 group. The SSVEP response generated by the lowest stimulation frequency (i.e., 6 Hz) was rescued with the intake of psychostimulant medication. Conclusions Impaired processing of rhythmic visual stimulation was evidenced in children with NF1 through measures of SSVEP responses. Those responses seem to be more reduced in children with NF1 who exhibit more symptoms of inattention and emotional/behavioral problems in their daily life. SSVEPs are potentially sensitive electrophysiological markers that could be included in future studies investigating the impact of medication on brain activity and cognitive functioning in children with NF1. Supplementary Information The online version contains supplementary material available at 10.1186/s11689-022-09452-y.
Collapse
|
8
|
Ottenhoff MJ, Dijkhuizen S, Ypelaar ACH, de Oude NL, Koekkoek SKE, Wang SSH, De Zeeuw CI, Elgersma Y, Boele HJ. Cerebellum-dependent associative learning is not impaired in a mouse model of neurofibromatosis type 1. Sci Rep 2022; 12:19041. [PMID: 36351971 PMCID: PMC9646701 DOI: 10.1038/s41598-022-21429-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 09/27/2022] [Indexed: 11/11/2022] Open
Abstract
Individuals with Neurofibromatosis type 1 (NF1) experience a high degree of motor problems. The cerebellum plays a pivotal role in motor functioning and the NF1 gene is highly expressed in cerebellar Purkinje cells. However, it is not well understood to what extent NF1 affects cerebellar functioning and how this relates to NF1 motor functioning. Therefore, we subjected global Nf1+/- mice to a cerebellum-dependent associative learning task, called Pavlovian eyeblink conditioning. Additionally, we assessed general motor function and muscle strength in Nf1+/- mice. To our surprise, we found that Nf1+/- mice showed a moderately increased learning rate of conditioned eyeblink responses, as well as improved accuracy in the adaptive timing of the eyeblink responses. Locomotion, balance, general motor function, and muscle strength were not affected in Nf1+/- mice. Together, our results support the view that cerebellar function in Nf1+/- mice is unimpaired.
Collapse
Affiliation(s)
- M J Ottenhoff
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
- The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, 3015GD, The Netherlands
| | - S Dijkhuizen
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
| | - A C H Ypelaar
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
| | - N L de Oude
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
| | - S K E Koekkoek
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
| | - S S-H Wang
- Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ, USA
| | - C I De Zeeuw
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
- Royal Academy of Arts and Sciences (KNAW), Netherlands Institute for Neuroscience, 1105 BA, Amsterdam, The Netherlands
| | - Y Elgersma
- The ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus Medical Center, Rotterdam, 3015GD, The Netherlands
- Department of Clinical Genetics, Erasmus MC, 3000 DR, Rotterdam, The Netherlands
| | - H J Boele
- Department of Neuroscience, Erasmus MC, 3000 DR, Rotterdam, The Netherlands.
- Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ, USA.
| |
Collapse
|
9
|
CRISPR-Cas9-generated mouse model of neurofibromatosis type 1. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00256-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
10
|
Chesnokova E, Beletskiy A, Kolosov P. The Role of Transposable Elements of the Human Genome in Neuronal Function and Pathology. Int J Mol Sci 2022; 23:5847. [PMID: 35628657 PMCID: PMC9148063 DOI: 10.3390/ijms23105847] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 12/13/2022] Open
Abstract
Transposable elements (TEs) have been extensively studied for decades. In recent years, the introduction of whole-genome and whole-transcriptome approaches, as well as single-cell resolution techniques, provided a breakthrough that uncovered TE involvement in host gene expression regulation underlying multiple normal and pathological processes. Of particular interest is increased TE activity in neuronal tissue, and specifically in the hippocampus, that was repeatedly demonstrated in multiple experiments. On the other hand, numerous neuropathologies are associated with TE dysregulation. Here, we provide a comprehensive review of literature about the role of TEs in neurons published over the last three decades. The first chapter of the present review describes known mechanisms of TE interaction with host genomes in general, with the focus on mammalian and human TEs; the second chapter provides examples of TE exaptation in normal neuronal tissue, including TE involvement in neuronal differentiation and plasticity; and the last chapter lists TE-related neuropathologies. We sought to provide specific molecular mechanisms of TE involvement in neuron-specific processes whenever possible; however, in many cases, only phenomenological reports were available. This underscores the importance of further studies in this area.
Collapse
Affiliation(s)
- Ekaterina Chesnokova
- Laboratory of Cellular Neurobiology of Learning, Institute of Higher Nervous Activity and Neurophysiology of the Russian Academy of Sciences, 117485 Moscow, Russia; (A.B.); (P.K.)
| | | | | |
Collapse
|
11
|
Rabab’h O, Gharaibeh A, Al-Ramadan A, Ismail M, Shah J. Pharmacological Approaches in Neurofibromatosis Type 1-Associated Nervous System Tumors. Cancers (Basel) 2021; 13:cancers13153880. [PMID: 34359780 PMCID: PMC8345673 DOI: 10.3390/cancers13153880] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 07/28/2021] [Accepted: 07/28/2021] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Neurofibromatosis type 1 (NF1) is a common cancer predisposition genetic disease that is associated with significant morbidity and mortality. In this literature review, we discuss the major pathways in the nervous system that are affected by NF1, tumors that are associated with NF1, drugs that target these pathways, and genetic models of NF1. We also summarize the latest updates from clinical trials that are evaluating pharmacological agents to treat these tumors and discuss the efforts that are being made to cure the disease in the future Abstract Neurofibromatosis type 1 is an autosomal dominant genetic disease and a common tumor predisposition syndrome that affects 1 in 3000 to 4000 patients in the USA. Although studies have been conducted to better understand and manage this disease, the underlying pathogenesis of neurofibromatosis type 1 has not been completely elucidated, and this disease is still associated with significant morbidity and mortality. Treatment options are limited to surgery with chemotherapy for tumors in cases of malignant transformation. In this review, we summarize the advances in the development of targeted pharmacological interventions for neurofibromatosis type 1 and related conditions.
Collapse
Affiliation(s)
- Omar Rabab’h
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
| | - Abeer Gharaibeh
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
- Insight Institute of Neurosurgery & Neuroscience, Flint, MI 48507, USA
- Insight Surgical Hospital, Warren, MI 48091, USA
| | - Ali Al-Ramadan
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
| | - Manar Ismail
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
| | - Jawad Shah
- Insight Research Institute, Flint, MI 48507, USA; (O.R.); (A.G.); (A.A.-R.); (M.I.)
- Center for Cognition and Neuroethics, University of Michigan-Flint, Flint, MI 48502, USA
- Insight Institute of Neurosurgery & Neuroscience, Flint, MI 48507, USA
- Insight Surgical Hospital, Warren, MI 48091, USA
- Department of Medicine, College of Human Medicine, Michigan State University, East Lansing, MI 48824, USA
- Correspondence:
| |
Collapse
|
12
|
Karalis V, Bateup HS. Current Approaches and Future Directions for the Treatment of mTORopathies. Dev Neurosci 2021; 43:143-158. [PMID: 33910214 PMCID: PMC8440338 DOI: 10.1159/000515672] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/13/2021] [Indexed: 11/19/2022] Open
Abstract
The mechanistic target of rapamycin (mTOR) is a kinase at the center of an evolutionarily conserved signaling pathway that orchestrates cell growth and metabolism. mTOR responds to an array of intra- and extracellular stimuli and in turn controls multiple cellular anabolic and catabolic processes. Aberrant mTOR activity is associated with numerous diseases, with particularly profound impact on the nervous system. mTOR is found in two protein complexes, mTOR complex 1 (mTORC1) and 2 (mTORC2), which are governed by different upstream regulators and have distinct cellular actions. Mutations in genes encoding for mTOR regulators result in a collection of neurodevelopmental disorders known as mTORopathies. While these disorders can affect multiple organs, neuropsychiatric conditions such as epilepsy, intellectual disability, and autism spectrum disorder have a major impact on quality of life. The neuropsychiatric aspects of mTORopathies have been particularly challenging to treat in a clinical setting. Current therapeutic approaches center on rapamycin and its analogs, drugs that are administered systemically to inhibit mTOR activity. While these drugs show some clinical efficacy, adverse side effects, incomplete suppression of mTOR targets, and lack of specificity for mTORC1 or mTORC2 may limit their utility. An increased understanding of the neurobiology of mTOR and the underlying molecular, cellular, and circuit mechanisms of mTOR-related disorders will facilitate the development of improved therapeutics. Animal models of mTORopathies have helped unravel the consequences of mTOR pathway mutations in specific brain cell types and developmental stages, revealing an array of disease-related phenotypes. In this review, we discuss current progress and potential future directions for the therapeutic treatment of mTORopathies with a focus on findings from genetic mouse models.
Collapse
Affiliation(s)
- Vasiliki Karalis
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
| | - Helen S Bateup
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, California, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| |
Collapse
|
13
|
Rahn RM, Weichselbaum CT, Gutmann DH, Dougherty JD, Maloney SE. Shared developmental gait disruptions across two mouse models of neurodevelopmental disorders. J Neurodev Disord 2021; 13:10. [PMID: 33743598 PMCID: PMC7980331 DOI: 10.1186/s11689-021-09359-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Accepted: 03/05/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Motor deficits such as abnormal gait are an underappreciated yet characteristic phenotype of many neurodevelopmental disorders (NDDs), including Williams Syndrome (WS) and Neurofibromatosis Type 1 (NF1). Compared to cognitive phenotypes, gait phenotypes are readily and comparably assessed in both humans and model organisms and are controlled by well-defined CNS circuits. Discovery of a common gait phenotype between NDDs might suggest shared cellular and molecular deficits and highlight simple outcome variables to potentially quantify longitudinal treatment efficacy in NDDs. METHODS We characterized gait using the DigiGait assay in two different murine NDD models: the complete deletion (CD) mouse, which models hemizygous loss of the complete WS locus, and the Nf1+/R681X mouse, which models a NF1 patient-derived heterozygous germline NF1 mutation. Longitudinal data were collected across four developmental time points (postnatal days 21-30) and one early adulthood time point. RESULTS Compared to wildtype littermate controls, both models displayed markedly similar spatial, temporal, and postural gait abnormalities during development. Developing CD mice also displayed significant decreases in variability metrics. Multiple gait abnormalities observed across development in the Nf1+/R681X mice persisted into early adulthood, including increased stride length and decreased stride frequency, while developmental abnormalities in the CD model largely resolved by adulthood. CONCLUSIONS These findings suggest that the subcomponents of gait affected in NDDs show overlap between disorders as well as some disorder-specific features, which may change over the course of development. Our incorporation of spatial, temporal, and postural gait measures also provides a template for gait characterization in other NDD models and a platform to examining circuits or longitudinal therapeutics.
Collapse
Affiliation(s)
- Rachel M Rahn
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Radiology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA
| | - Claire T Weichselbaum
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA
| | - David H Gutmann
- Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA
| | - Susan E Maloney
- Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA. .,Intellectual and Developmental Disabilities Research Center, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, MO, 63110-1093, USA.
| |
Collapse
|
14
|
Fountain DM, Smith MJ, O'Leary C, Pathmanaban ON, Roncaroli F, Bobola N, King AT, Evans DG. The spatial phenotype of genotypically distinct meningiomas demonstrate potential implications of the embryology of the meninges. Oncogene 2021; 40:875-884. [PMID: 33262459 PMCID: PMC8440207 DOI: 10.1038/s41388-020-01568-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/05/2020] [Accepted: 11/11/2020] [Indexed: 12/29/2022]
Abstract
Meningiomas are the most common primary brain tumor and their incidence and prevalence is increasing. This review summarizes current evidence regarding the embryogenesis of the human meninges in the context of meningioma pathogenesis and anatomical distribution. Though not mutually exclusive, chromosomal instability and pathogenic variants affecting the long arm of chromosome 22 (22q) result in meningiomas in neural-crest cell-derived meninges, while variants affecting Hedgehog signaling, PI3K signaling, TRAF7, KLF4, and POLR2A result in meningiomas in the mesodermal-derived meninges of the midline and paramedian anterior, central, and ventral posterior skull base. Current evidence regarding the common pathways for genetic pathogenesis and the anatomical distribution of meningiomas is presented alongside existing understanding of the embryological origins for the meninges prior to proposing next steps for this work.
Collapse
Affiliation(s)
- Daniel M Fountain
- Geoffrey Jefferson Brain Research Centre, Salford Royal NHS Foundation Trust and the University of Manchester, Manchester, UK.
| | - Miriam J Smith
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre (MAHSC), St Mary's Hospital, School of Biological Sciences, Division of Evolution and Genomic Sciences, University of Manchester, Manchester, UK
| | - Claire O'Leary
- Geoffrey Jefferson Brain Research Centre, Salford Royal NHS Foundation Trust and the University of Manchester, Manchester, UK
| | - Omar N Pathmanaban
- Geoffrey Jefferson Brain Research Centre, Salford Royal NHS Foundation Trust and the University of Manchester, Manchester, UK
| | - Federico Roncaroli
- Geoffrey Jefferson Brain Research Centre, Salford Royal NHS Foundation Trust and the University of Manchester, Manchester, UK
| | - Nicoletta Bobola
- School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Andrew T King
- Geoffrey Jefferson Brain Research Centre, Salford Royal NHS Foundation Trust and the University of Manchester, Manchester, UK
| | - Dafydd Gareth Evans
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre (MAHSC), St Mary's Hospital, School of Biological Sciences, Division of Evolution and Genomic Sciences, University of Manchester, Manchester, UK
| |
Collapse
|
15
|
Nix JS, Blakeley J, Rodriguez FJ. An update on the central nervous system manifestations of neurofibromatosis type 1. Acta Neuropathol 2020; 139:625-641. [PMID: 30963251 DOI: 10.1007/s00401-019-02002-2] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Revised: 03/30/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Neurofibromatosis 1 (NF1) is an autosomal dominant genetic disorder that presents with variable phenotypes as a result of mutations in the neurofibromatosis type 1 (NF1) gene and subsequently, abnormal function of the protein product, neurofibromin. Patients with NF1 are at increased risk for central nervous system (CNS) manifestations including structural, functional, and neoplastic disease. The mechanisms underlying the varied manifestations of NF1 are incompletely understood, but the loss of functional neurofibromin, resulting in sustained activation of the oncoprotein RAS, is responsible for tumorigenesis throughout the body, including the CNS. Much of our understanding of NF1-related CNS manifestations is from a combination of data from animal models and natural history studies of people with NF1 and CNS disease. Data from animal models suggest the importance of both Nf1 mutations and somatic genetic alterations, such as Tp53 loss, for development of neoplasms, as well as the role of the timing of the acquisition of such alterations on the variability of CNS manifestations. A variety of non-neoplastic structural (macrocephaly, hydrocephalus, aqueductal stenosis, and vasculopathy) and functional (epilepsy, impaired cognition, attention deficits, and autism spectrum disorder) abnormalities occur with variable frequency in individuals with NF1. In addition, there is increasing evidence that similar appearing CNS neoplasms in people with and without the NF1 syndrome are due to distinct oncogenic pathways. Gliomas in people with NF1 show alterations in the RAS/MAPK pathway, generally in the absence of BRAF alterations (common to sporadic pilocytic astrocytomas) or IDH or histone H3 mutations (common to diffuse gliomas subsets). A subset of low-grade astrocytomas in these patients remain difficult to classify using standard criteria, and occasionally demonstrate morphologic features resembling subependymal giant cell astrocytomas that afflict patients with tuberous sclerosis complex ("SEGA-like astrocytomas"). There is also emerging evidence that NF1-associated high-grade astrocytomas have frequent co-existing alterations such as ATRX mutations and an alternative lengthening of telomeres (ALT) phenotype responsible for unique biologic properties. Ongoing efforts are seeking to improve diagnostic accuracy for CNS neoplasms in the setting of NF1 versus sporadic tumors. In addition, MEK inhibitors, which act on the RAS/MAPK pathway, continue to be studied as rational targets for the treatment of NF1-associated tumors, including CNS tumors.
Collapse
|
16
|
Towards a neurobiological understanding of pain in neurofibromatosis type 1: mechanisms and implications for treatment. Pain 2020; 160:1007-1018. [PMID: 31009417 DOI: 10.1097/j.pain.0000000000001486] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Neurofibromatosis type 1 (NF1) is the most common of a group of rare diseases known by the term, "Neurofibromatosis," affecting 1 in 3000 to 4000 people. NF1 patients present with, among other disease complications, café au lait patches, skin fold freckling, Lisch nodules, orthopedic complications, cutaneous neurofibromas, malignant peripheral nerve sheath tumors, cognitive impairment, and chronic pain. Although NF1 patients inevitably express pain as a debilitating symptom of the disease, not much is known about its manifestation in the NF1 disease, with most current information coming from sporadic case reports. Although these reports indicate the existence of pain, the molecular signaling underlying this symptom remains underexplored, and thus, we include a synopsis of the literature surrounding NF1 pain studies in 3 animal models: mouse, rat, and miniswine. We also highlight unexplored areas of NF1 pain research. As therapy for NF1 pain remains in various clinical and preclinical stages, we present current treatments available for patients and highlight the importance of future therapeutic development. Equally important, NF1 pain is accompanied by psychological complications in comorbidities with sleep, gastrointestinal complications, and overall quality of life, lending to the importance of investigation into this understudied phenomenon of NF1. In this review, we dissect the presence of pain in NF1 in terms of psychological implication, anatomical presence, and discuss mechanisms underlying the onset and potentiation of NF1 pain to evaluate current therapies and propose implications for treatment of this severely understudied, but prevalent symptom of this rare disease.
Collapse
|
17
|
A cerebellopontine angle mouse model for the investigation of tumor biology, hearing, and neurological function in NF2-related vestibular schwannoma. Nat Protoc 2019; 14:541-555. [PMID: 30617350 DOI: 10.1038/s41596-018-0105-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Neurofibromatosis type II (NF2) is a disease that lacks effective therapies. NF2 is characterized by bilateral vestibular schwannomas (VSs) that cause progressive and debilitating hearing loss, leading to social isolation and increased rates of depression. A major limitation in NF2 basic and translational research is the lack of animal models that allow the full spectrum of research into the biology and molecular mechanisms of NF2 tumor progression, as well as the effects on neurological function. In this protocol, we describe how to inject schwannoma cells into the mouse brain cerebellopontine angle (CPA) region. We also describe how to apply state-of-the-art intravital imaging and hearing assessment techniques to study tumor growth and hearing loss. In addition, ataxia, angiogenesis, and tumor-stroma interaction assays can be applied, and the model can be used to test the efficacy of novel therapeutic approaches. By studying the disease from every angle, this model offers the potential to unravel the basic biological underpinnings of NF2 and to develop novel therapeutics to control this devastating disease. Our protocol can be adapted to study other diseases within the CPA, including meningiomas, lipomas, vascular malformations, hemangiomas, epidermoid cysts, cerebellar astrocytomas, and metastatic lesions. The entire surgical procedure takes ~45 min per mouse and allows for subsequent longitudinal imaging, as well as neurological and hearing assessment, for up to 2 months.
Collapse
|
18
|
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: 55] [Impact Index Per Article: 7.9] [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
|
19
|
White KA, Swier VJ, Cain JT, Kohlmeyer JL, Meyerholz DK, Tanas MR, Uthoff J, Hammond E, Li H, Rohret FA, Goeken A, Chan CH, Leidinger MR, Umesalma S, Wallace MR, Dodd RD, Panzer K, Tang AH, Darbro BW, Moutal A, Cai S, Li W, Bellampalli SS, Khanna R, Rogers CS, Sieren JC, Quelle DE, Weimer JM. A porcine model of neurofibromatosis type 1 that mimics the human disease. JCI Insight 2018; 3:120402. [PMID: 29925695 DOI: 10.1172/jci.insight.120402] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/17/2018] [Indexed: 12/11/2022] Open
Abstract
Loss of the NF1 tumor suppressor gene causes the autosomal dominant condition, neurofibromatosis type 1 (NF1). Children and adults with NF1 suffer from pathologies including benign and malignant tumors to cognitive deficits, seizures, growth abnormalities, and peripheral neuropathies. NF1 encodes neurofibromin, a Ras-GTPase activating protein, and NF1 mutations result in hyperactivated Ras signaling in patients. Existing NF1 mutant mice mimic individual aspects of NF1, but none comprehensively models the disease. We describe a potentially novel Yucatan miniswine model bearing a heterozygotic mutation in NF1 (exon 42 deletion) orthologous to a mutation found in NF1 patients. NF1+/ex42del miniswine phenocopy the wide range of manifestations seen in NF1 patients, including café au lait spots, neurofibromas, axillary freckling, and neurological defects in learning and memory. Molecular analyses verified reduced neurofibromin expression in swine NF1+/ex42del fibroblasts, as well as hyperactivation of Ras, as measured by increased expression of its downstream effectors, phosphorylated ERK1/2, SIAH, and the checkpoint regulators p53 and p21. Consistent with altered pain signaling in NF1, dysregulation of calcium and sodium channels was observed in dorsal root ganglia expressing mutant NF1. Thus, these NF1+/ex42del miniswine recapitulate the disease and provide a unique, much-needed tool to advance the study and treatment of NF1.
Collapse
Affiliation(s)
- Katherine A White
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Vicki J Swier
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | - Jacob T Cain
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | | | | | | | - Johanna Uthoff
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Biomedical Engineering at the University of Iowa, Iowa City, Iowa, USA
| | - Emily Hammond
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Biomedical Engineering at the University of Iowa, Iowa City, Iowa, USA
| | - Hua Li
- Department of Molecular Genetics and Microbiology and.,University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | | | | | - Chun-Hung Chan
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA
| | | | | | - Margaret R Wallace
- Department of Molecular Genetics and Microbiology and.,University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Rebecca D Dodd
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Karin Panzer
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Amy H Tang
- Department of Microbiology and Molecular Cell Biology, Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, Virginia
| | - Benjamin W Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA.,Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA
| | - Aubin Moutal
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Song Cai
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | - Wennan Li
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | | | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, Arizona, USA
| | | | - Jessica C Sieren
- Department of Radiology, University of Iowa Carver College of Medicine, Iowa City, Iowa, USA.,Department of Biomedical Engineering at the University of Iowa, Iowa City, Iowa, USA.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Dawn E Quelle
- Molecular Medicine Program.,Department of Pathology, and.,Department of Pharmacology and.,Holden Comprehensive Cancer Center, University of Iowa, Iowa City, Iowa, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, South Dakota, USA.,Department of Pediatrics, Sanford School of Medicine at the University of South Dakota, Sioux Falls, South Dakota, USA
| |
Collapse
|
20
|
Dalal S, Connelly B, Singh M, Singh K. NF2 signaling pathway plays a pro-apoptotic role in β-adrenergic receptor stimulated cardiac myocyte apoptosis. PLoS One 2018; 13:e0196626. [PMID: 29709009 PMCID: PMC5927447 DOI: 10.1371/journal.pone.0196626] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Accepted: 04/16/2018] [Indexed: 12/24/2022] Open
Abstract
β-adrenergic receptor (β-AR) stimulation induces cardiac myocyte apoptosis in vitro and in vivo. Neurofibromin 2 (NF2) is a member of the ezrin/radixin/moesin (ERM) family of proteins. Post-translational modifications such as phosphorylation and sumoylation affect NF2 activity, subcellular localization and function. Here, we tested the hypothesis that β-AR stimulation induces post-translational modifications of NF2, and NF2 plays a pro-apoptotic role in β-AR-stimulated myocyte apoptosis.
Collapse
Affiliation(s)
- Suman Dalal
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Barbara Connelly
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Mahipal Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
| | - Krishna Singh
- Department of Biomedical Sciences, James H Quillen College of Medicine, East Tennessee State University, Johnson City, TN, United States of America
- Center for Inflammation, Infectious Disease and Immunity, East Tennessee State University, Johnson City, TN, United States of America
- James H Quillen Veterans Affairs Medical Center, Mountain Home, TN, United States of America
- * E-mail:
| |
Collapse
|
21
|
Maloney SE, Chandler KC, Anastasaki C, Rieger MA, Gutmann DH, Dougherty JD. Characterization of early communicative behavior in mouse models of neurofibromatosis type 1. Autism Res 2017; 11:44-58. [PMID: 28842941 DOI: 10.1002/aur.1853] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Revised: 06/26/2017] [Accepted: 07/23/2017] [Indexed: 01/23/2023]
Abstract
Neurofibromatosis type 1 (NF1) is a monogenic neurodevelopmental disease caused by germline loss-of-function mutations in the NF1 tumor suppressor gene. Cognitive impairments are observed in approximately 80% of children with this disease, with 45-60% exhibiting autism spectrum disorder (ASD) symptomatology. In light of the high comorbidity rate between ASD and NF1, we assessed early communicative behavior by maternal-separation induced pup ultrasonic vocalizations (USV) and developmental milestones in two distinct Nf1 genetically engineered models, one modeling clinical germline heterozygous loss of Nf1 function (Nf1+/- mice), and a second with somatic biallelic Nf1 inactivation in neuroglial progenitor cells (Nf1GFAP CKO mice). We observed altered USV production in both models: Nf1+/- mice exhibited both increased USVs across development and alterations in aspects of pitch, while Nf1GFAP CKO mice demonstrated a decrease in USVs. Developmental milestones, such as weight, pinnae detachment, and eye opening, were not disrupted in either model, indicating the USV deficits were not due to gross developmental delay, and likely reflected more specific alterations in USV circuitry. In this respect, increased whole-brain serotonin was observed in Nf1+/- mice, but whole-brain levels of dopamine and its metabolites were unchanged at the age of peak USV disruption, and USV alterations did not correlate with overall level of neurofibromin loss. The early communicative phenotypes reported herein should motivate further studies into the risks mediated by haploinsufficiency and biallelic deletion of Nf1 across a full battery of ASD-relevant behavioral phenotypes, and a targeted analysis of underlying circuitry disruptions. Autism Res 2018, 11: 44-58. © 2017 International Society for Autism Research, Wiley Periodicals, Inc. LAY SUMMARY Neurofibromatosis type 1 (NF1) is a common neurogenetic disorder caused by mutation of the NF1 gene, in which 80% of affected children exhibit cognitive and behavioral issues. Based on emerging evidence that NF1 may be an autism predisposition gene, we examined autism spectrum disorder (ASD)-relevant early communicative behavior in Nf1 mouse models and observed alterations in both models. The changes in early communicative behavior in Nf1 mutant mice should motivate further studies into the causative factors and potential treatments for ASD arising in the context of NF1.
Collapse
Affiliation(s)
- Susan E Maloney
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri.,Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri
| | - Krystal C Chandler
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri.,Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri
| | - Corina Anastasaki
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri
| | - Michael A Rieger
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri.,Department of Psychiatry, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri
| | - Joseph D Dougherty
- Department of Genetics, Washington University School of Medicine, 660 South Euclid Avenue, St. Louis, Missouri
| |
Collapse
|
22
|
Yoshizawa K, Yuki M, Kinoshita Y, Emoto Y, Yuri T, Elmore SA, Tsubura A. N-methyl-N-nitrosourea-induced schwannomas in male Sprague-Dawley rats with a literature review of inducible and spontaneous lesions. ACTA ACUST UNITED AC 2016; 68:371-9. [PMID: 27233116 DOI: 10.1016/j.etp.2016.05.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/03/2016] [Accepted: 05/10/2016] [Indexed: 12/01/2022]
Abstract
N-methyl-N-nitrosourea (MNU) possesses peripheral nervous system carcinogenic activity in rats and induces benign and malignant schwannomas in systemic organs. In this retrospective study, we compared the characteristics of various immunohistochemical markers in MNU-induced schwannomas in male Crj:CD(SD)IGS rats including: vimentin (Vim), S100, p75 nerve growth factor receptor (LNGFR), CD57, pancytokeratin (CK), myoglobin, desmin and α smooth muscle actin (SMA). Single intraperitoneal exposures of 50 or 75mg/kg MNU in male rats at the age of 4 weeks induced schwannomas in 43 surviving and terminated rats up to 30-weeks-old. The incidence rate of neoplastic lesions was 37% (16 of 43 rats). Benign schwannomas (mesentery, pancreas, thymus) and malignant schwannomas (subendocardium, cardiac intramural, thoracic cavity, abdominal cavity, prostate), occurred in nine and seven rats, respectively. All neoplastic lesions were moderately or strongly positive for Vim, S100 and LNGFR proteins. Benign tumors were weakly positive and malignant tumors strongly positive for Ki-67, suggesting a high active proliferation rate of Schwann cell precursors. All lesions were negative for CD57, CK, myoglobin, desmin and SMA. This data may provide useful immunohistochemical information for the investigation of schwannomas in rat chemical carcinogenicity studies.
Collapse
Affiliation(s)
- Katsuhiko Yoshizawa
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan.
| | - Michiko Yuki
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Yuichi Kinoshita
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan; Division of Diagnostic Cytopathology and Histopathology, Kansai Medical University Medical Center, Fumizono 10-15, Moriguchi, Osaka 570-8507, Japan
| | - Yuko Emoto
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Takashi Yuri
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| | - Susan A Elmore
- Cellular & Molecular Pathology Branch, National Institute of Environmental Health Sciences, 111 T.W. Alexander Drive, Research Triangle Park, NC 27709, USA
| | - Airo Tsubura
- Department of Pathology II, Kansai Medical University, 2-5-1 Shin-machi, Hirakata, Osaka 573-1010, Japan
| |
Collapse
|
23
|
Watson AL, Carlson DF, Largaespada DA, Hackett PB, Fahrenkrug SC. Engineered Swine Models of Cancer. Front Genet 2016; 7:78. [PMID: 27242889 PMCID: PMC4860525 DOI: 10.3389/fgene.2016.00078] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
Over the past decade, the technology to engineer genetically modified swine has seen many advancements, and because their physiology is remarkably similar to that of humans, swine models of cancer may be extremely valuable for preclinical safety studies as well as toxicity testing of pharmaceuticals prior to the start of human clinical trials. Hence, the benefits of using swine as a large animal model in cancer research and the potential applications and future opportunities of utilizing pigs in cancer modeling are immense. In this review, we discuss how pigs have been and can be used as a biomedical models for cancer research, with an emphasis on current technologies. We have focused on applications of precision genetics that can provide models that mimic human cancer predisposition syndromes. In particular, we describe the advantages of targeted gene-editing using custom endonucleases, specifically TALENs and CRISPRs, and transposon systems, to make novel pig models of cancer with broad preclinical applications.
Collapse
Affiliation(s)
| | | | - David A Largaespada
- RecombineticsSt. Paul, MN, USA; Masonic Cancer Center, University of MinnesotaMinneapolis, MN, USA; Genetics, Cell Biology and Development, University of MinnesotaMinneapolis, MN, USA; Pediatrics, University of MinnesotaMinneapolis, MN, USA
| | - Perry B Hackett
- RecombineticsSt. Paul, MN, USA; Genetics, Cell Biology and Development, University of MinnesotaMinneapolis, MN, USA; Center for Genome Engineering, University of MinnesotaMinneapolis, MN, USA
| | | |
Collapse
|
24
|
Abstract
Neurofibromatosis type 1 (NF1) is a relatively common tumour predisposition syndrome related to germline aberrations of NF1, a tumour suppressor gene. The gene product neurofibromin is a negative regulator of the Ras cellular proliferation pathway, and also exerts tumour suppression via other mechanisms. Recent next-generation sequencing projects have revealed somatic NF1 aberrations in various sporadic tumours. NF1 plays a critical role in a wide range of tumours. NF1 alterations appear to be associated with resistance to therapy and adverse outcomes in several tumour types. Identification of a patient's germline or somatic NF1 aberrations can be challenging, as NF1 is one of the largest human genes, with a myriad of possible mutations. Epigenetic factors may also contribute to inadequate levels of neurofibromin in cancer cells. Clinical trials of NF1-based therapeutic approaches are currently limited. Preclinical studies on neurofibromin-deficient malignancies have mainly been on malignant peripheral nerve sheath tumour cell lines or xenografts derived from NF1 patients. However, the emerging recognition of the role of NF1 in sporadic cancers may lead to the development of NF1-based treatments for other tumour types. Improved understanding of the implications of NF1 aberrations is critical for the development of novel therapeutic strategies.
Collapse
|
25
|
Anti-VEGF treatment improves neurological function and augments radiation response in NF2 schwannoma model. Proc Natl Acad Sci U S A 2015; 112:14676-81. [PMID: 26554010 DOI: 10.1073/pnas.1512570112] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Hearing loss is the main limitation of radiation therapy for vestibular schwannoma (VS), and identifying treatment options that minimize hearing loss are urgently needed. Treatment with bevacizumab is associated with tumor control and hearing improvement in neurofibromatosis type 2 (NF2) patients; however, its effect is not durable and its mechanism of action on nerve function is unknown. We modeled the effect anti-VEGF therapy on neurological function in the sciatic nerve model and found that it improves neurological function by alleviating tumor edema, which may further improve results by decreasing muscle atrophy and increasing nerve regeneration. Using a cranial window model, we showed that anti-VEGF treatment may achieve these effects via normalizing the tumor vasculature, improving vessel perfusion, and delivery of oxygenation. It is known that oxygen is a potent radiosensitizer; therefore, we further demonstrated that combining anti-VEGF with radiation therapy can achieve a better tumor control and help lower the radiation dose and, thus, minimize radiation-related neurological toxicity. Our results provide compelling rationale for testing combined therapy in human VS.
Collapse
|
26
|
Apostolova I, Niedzielska D, Derlin T, Koziolek EJ, Amthauer H, Salmen B, Pahnke J, Brenner W, Mautner VF, Buchert R. Perfusion single photon emission computed tomography in a mouse model of neurofibromatosis type 1: towards a biomarker of neurologic deficits. J Cereb Blood Flow Metab 2015; 35:1304-12. [PMID: 25785829 PMCID: PMC4528004 DOI: 10.1038/jcbfm.2015.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2014] [Revised: 01/21/2015] [Accepted: 02/16/2015] [Indexed: 12/16/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a single-gene disorder affecting neurologic function in humans. The NF1+/- mouse model with germline mutation of the NF1 gene presents with deficits in learning, attention, and motor coordination, very similar to NF1 patients. The present study performed brain perfusion single-photon emission computed tomography (SPECT) in NF1+/- mice to identify possible perfusion differences as surrogate marker for altered cerebral activity in NF1. Cerebral perfusion was measured with hexamethyl-propyleneamine oxime (HMPAO) SPECT in NF1+/- mice and their wild-type littermates longitudinally at juvenile age and at young adulthood. Histology and immunohistochemistry were performed to test for structural changes. There was increased HMPAO uptake in NF1 mice in the amygdala at juvenile age, which reduced to normal levels at young adulthood. There was no genotype effect on thalamic HMPAO uptake, which was confirmed by ex vivo measurements of F-18-fluorodeoxyglucose uptake in the thalamus. Morphologic analyses showed no major structural abnormalities. However, there was some evidence of increased density of microglial somata in the amygdala of NF1-deficient mice. In conclusion, there is evidence of increased perfusion and increased density of microglia in juvenile NF1 mice specifically in the amygdala, both of which might be associated with altered synaptic plasticity and, therefore, with cognitive deficits in NF1.
Collapse
Affiliation(s)
- Ivayla Apostolova
- 1] Department of Nuclear Medicine, University Medicine Charité Berlin, Berlin, Germany [2] Department of Radiology and Nuclear Medicine, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - Dagmara Niedzielska
- Department of Nuclear Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Eva J Koziolek
- Department of Diagnostic and Interventional Radiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Holger Amthauer
- Department of Radiology and Nuclear Medicine, University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - Benedikt Salmen
- Neuroscience Research Center, University Medicine Charité Berlin, Berlin, Germany
| | - Jens Pahnke
- 1] Department of Neuropathology, Oslo University Hospital (OUS), University of Oslo (UiO), Oslo, Norway [2] LIED, University of Lübeck, Lübeck, Germany [3] Leibniz Institute of Plant Biochemistry, Halle, Germany
| | - Winfried Brenner
- Department of Nuclear Medicine, University Medicine Charité Berlin, Berlin, Germany
| | - Victor F Mautner
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ralph Buchert
- Department of Nuclear Medicine, University Medicine Charité Berlin, Berlin, Germany
| |
Collapse
|
27
|
Ahmad I, Yue WY, Fernando A, Clark JJ, Woodson EA, Hansen MR. p75NTR is highly expressed in vestibular schwannomas and promotes cell survival by activating nuclear transcription factor κB. Glia 2014; 62:1699-712. [PMID: 24976126 PMCID: PMC4150679 DOI: 10.1002/glia.22709] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 06/03/2014] [Accepted: 06/06/2014] [Indexed: 01/08/2023]
Abstract
Vestibular schwannomas (VSs) arise from Schwann cells (SCs) and result from the loss of function of merlin, the protein product of the NF2 tumor suppressor gene. In contrast to non-neoplastic SCs, VS cells survive long-term in the absence of axons. We find that p75(NTR) is overexpressed in VSs compared with normal nerves, both at the transcript and protein level, similar to the response of non-neoplastic SCs following axotomy. Despite elevated p75(NTR) expression, VS cells are resistant to apoptosis due to treatment with proNGF, a high affinity ligand for p75(NTR) . Furthermore, treatment with proNGF protects VS cells from apoptosis due to c-Jun N-terminal kinase (JNK) inhibition indicating that p75(NTR) promotes VS cell survival. Treatment of VS cells with proNGF activated NF-κB while inhibition of JNK with SP600125 or siRNA-mediated knockdown reduced NF-κB activity. Significantly, proNGF also activated NF-κB in cultures treated with JNK inhibitors. Thus, JNK activity appears to be required for basal levels of NF-κB activity but not for proNGF-induced NF-κB activity. To confirm that the increase in NF-κB activity contributes to the prosurvival effect of proNGF, we infected VS cultures with Ad.IκB.SerS32/36A virus, which inhibits NF-κB activation. Compared with control virus, Ad.IκB.SerS32/36A significantly increased apoptosis including in VS cells treated with proNGF. Thus, in contrast to non-neoplastic SCs, p75(NTR) signaling provides a prosurvival response in VS cells by activating NF-κB independent of JNK. Such differences may contribute to the ability of VS cells to survive long-term in the absence of axons.
Collapse
Affiliation(s)
- Iram Ahmad
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
| | - Wei Ying Yue
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
- Department of Otolaryngology-HNS, Mayo Clinic, Rochester, MN
| | - Augusta Fernando
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
- Department of Otolaryngology-HNS, Northwestern University, Chicago, IL
| | - J. Jason Clark
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
| | - Erika A. Woodson
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
- Department of Otolaryngology-HNS, Cleveland Clinic, Cleveland, OH
| | - Marlan R. Hansen
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242
| |
Collapse
|
28
|
Lin AL, Gutmann DH. Advances in the treatment of neurofibromatosis-associated tumours. Nat Rev Clin Oncol 2013; 10:616-24. [DOI: 10.1038/nrclinonc.2013.144] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
|
29
|
Spitsbergen JM, Frattini SA, Bowser PR, Getchell RG, Coffee LL, Wolfe MJ, Fisher JP, Marinovic SJ, Harr KE. Epizootic neoplasia of the lateral line system of lake trout (Salvelinus namaycush) in New York's Finger Lakes. Vet Pathol 2013; 50:418-33. [PMID: 23528941 DOI: 10.1177/0300985813482949] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This article documents an epizootic of inflammation and neoplasia selectively affecting the lateral line system of lake trout (Salvelinus namaycush) in 4 Finger Lakes in New York from 1985 to 1994. We studied more than 100 cases of this disease. Tumors occurred in 8% (5/64) of mature and 21% (3/14) of immature lake trout in the most severely affected lake. Lesions consisted of 1 or more neoplasm(s) in association with lymphocytic inflammation, multifocal erosions, and ulcerations of the epidermis along the lateral line. Lesions progressed from inflammatory to neoplastic, with 2-year-old lake trout showing locally extensive, intense lymphocytic infiltrates; 2- to 3-year-old fish having multiple, variably sized white masses up to 3 mm in diameter; and fish over 5 years old exhibiting 1 or more white, cerebriform masses greater than 1 cm in diameter. Histologic diagnoses of the tumors were predominantly spindle cell sarcomas or benign or malignant peripheral nerve sheath neoplasms, with fewer epitheliomas and carcinomas. Prevalence estimates did not vary significantly between sexes or season. The cause of this epizootic remains unclear. Tumor transmission trials, virus isolation procedures, and ultrastructural study of lesions failed to reveal evidence of a viral etiology. The Finger Lakes in which the disease occurred did not receive substantially more chemical pollution than unaffected lakes in the same chain during the epizootic, making an environmental carcinogen an unlikely primary cause of the epizootic. A hereditary component, however, may have contributed to this syndrome since only fish of the Seneca Lake strain were affected.
Collapse
Affiliation(s)
- J M Spitsbergen
- Department of Microbiology, 220 Nash Hall, Oregon State University, Corvallis, OR 97331, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Abstract
Much progress has been made in understanding the molecular genetics of brain tumors, especially gliomas.The development and use of high-throughput platforms that can interrogate molecular lesions on a variety of platforms will increase our ability to identify molecular subclasses of these tumors. Future challenges will include the development of methods to integrate these data among different platforms in order to identify optimal biomarkers and robust subclasses. The ultimate challenge, however, remains the translation of this biological knowledge into improved therapies for patients.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiation Oncology, University of Texas-MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
31
|
Rehg JE, Ward JM. Morphological and Immunohistochemical Characterization of Sarcomatous Tumors in Wild-Type and Genetically Engineered Mice. Vet Pathol 2011; 49:206-17. [DOI: 10.1177/0300985811429813] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Malignant soft tissue tumors are commonly observed in wild-type and gene-targeted mice. These tumors have different degrees of differentiation, cellularity, cellular atypia, nuclear pleomorphism, normal and abnormal mitosis, and giant tumor cells with enlarged polylobulated nuclei. They are often diagnosed as pleomorphic sarcoma, undifferentiated sarcoma, fibrosarcoma, malignant fibrous histiocytoma, sarcoma, or sarcoma, not otherwise specified. Pleomorphic sarcomas have no morphological differentiation toward a differentiated mesenchymal or other tumor type in hematoxylin and eosin–stained sections. With the use of immunohistochemistry, human and mouse, tumors associated with these broad nonspecific diagnoses can often be demonstrated to be of a specific cellular lineage. With mouse models being used to delineate the molecular mechanisms, pathogenesis, and cellular origin of human sarcomas, it will be necessary to correlate the morphological and cellular lineage and the molecular profiles of the pleomorphic tumors associated with these mouse models. The results presented here show that with the use of immunohistochemistry, the cellular lineage of many mouse tumors with pleomorphic features can be determined.
Collapse
Affiliation(s)
- J. E. Rehg
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, Tennessee
| | - J. M. Ward
- Global Vet Pathology, Montgomery Village, Maryland
| |
Collapse
|
32
|
Tucker T, Riccardi VM, Brown C, Fee J, Sutcliffe M, Vielkind J, Wechsler J, Wolkenstein P, Friedman JM. S100B and neurofibromin immunostaining and X-inactivation patterns of laser-microdissected cells indicate a multicellular origin of some NF1-associated neurofibromas. J Neurosci Res 2011; 89:1451-60. [PMID: 21674567 DOI: 10.1002/jnr.22654] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2010] [Revised: 02/22/2011] [Accepted: 03/03/2011] [Indexed: 01/10/2023]
Abstract
Neurofibromatosis 1 (NF1) is an autosomal dominant disease that predisposes individuals to developing benign neurofibromas. Some features and consequences of NF1 appear to result from partial deficiency of neurofibromin (Nfn), the NF1 gene protein product, as a result of haploinsufficiency for the NF1 gene. Other features and consequences of NF1 appear to involve total deficiency of Nfn, which arises as a result of either loss of function of the second NF1 allele or excess degradation of Nfn produced by the second allele in a particular clone of cells. We used immunofluorescence to assess the presence of Nfn in putative Schwann cells (S100B(+) ) and non-Schwann cells (S100B(-) ) in 36 NF1-derived benign neurofibromas classified histologically as diffuse or encapsulated. The S100B(+) /Nfn(-) cell population made up only 18% ± 10% (mean ± standard deviation) of the neurofibroma cells in both the diffuse and encapsulated neurofibromas. The proportion of S100B(+) /Nfn(+) cells was significantly higher and the proportion of S100B(-) /Nfn(-) cells was significantly lower in diffuse neurofibromas than in encapsulated neurofibromas. We isolated S100B(+) /Nfn(+) , S100B(+) /Nfn(-) , and S100B(-) /Nfn(+) cells by laser microdissection and, using X-chromosome inactivation profiles, assessed clonality for each cell type. We showed that, although some neurofibromas include a subpopulation of S100B(+) /Nfn(-) cells consistent with clonal expansion of a Schwann cell progenitor that has lost function of both NF1 alleles, other neurofibromas do not show evidence of monoclonal proliferation of Schwann cells. Our findings suggest that, although clonal loss of neurofibromin function is probably involved in the development of some NF1-associated neurofibromas, other pathogenic processes also occur.
Collapse
Affiliation(s)
- Tracy Tucker
- Department of Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The skin plays a critical role in the detection of internal malignances. Cutaneous signs of these disorders afford clinicians opportunities for early diagnosis and treatment. We aim to succinctly review the recognition, diagnosis, and treatment of selected cutaneous paraneoplastic diseases. Skin disorders that may be associated with paraneoplastic syndromes include: cutaneous metastases, tripe palms, Sweet's syndrome, glucagonoma, Paget's disease and extramammary Paget's disease, acanthosis nigricans, Birt-Hogg-Dube syndrome, basal cell nevus syndrome, Bazex syndrome (acrokeratosis paraneoplastica), carcinoid syndrome, Cowden's disease(multiple hamartoma syndrome), dermatomyositis, erythema gyratum repens, ichthyosis aquisita, von Recklinghausen's disease, pityriasis rotunda, pyoderma gangrenosum, Quincke's edema (angioedema and paraneoplastic uricaria), paraneoplastic pemphigus, Degos' disease, superior vena cava syndrome, Werner's syndrome, diffuse normolipemic plane xanthomas, and yellow nail syndrome. Treatment for these disorders depends on the nature and anatomic distribution of the primary neoplastic process.
Collapse
|
34
|
Abstract
Animal models of cancer have been instrumental in understanding the progression and therapy of hereditary cancer syndromes. The ability to alter the genome of an individual mouse cell in both constitutive and inducible approaches has led to many novel insights into their human counterparts. In this review, knockout mouse models of inherited human cancer syndromes are presented and insights from the study of these models are highlighted.
Collapse
Affiliation(s)
- Sohail Jahid
- Departments of Medicine and Genetic Medicine, Weill Cornell College of Medicine, Cornell University, New York NY
| | - Steven Lipkin
- Departments of Medicine and Genetic Medicine, Weill Cornell College of Medicine, Cornell University, New York NY
| |
Collapse
|
35
|
Alves DA, Bell TM, Benton C, Rushing EJ, Stevens EL. Giant thoracic schwannoma in a rhesus macaque (Macaca mulatta). JOURNAL OF THE AMERICAN ASSOCIATION FOR LABORATORY ANIMAL SCIENCE : JAALAS 2010; 49:868-872. [PMID: 21205456 PMCID: PMC2994058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/25/2010] [Revised: 04/19/2010] [Accepted: 04/30/2010] [Indexed: 05/30/2023]
Abstract
A 15-y-old male rhesus macaque with a 3-d history of labored breathing, was culled from a nonhuman primate research colony after thoracic radiographs and exploratory surgery revealed a 10-cm, well-circumscribed space-occupying mass in the posterior thoracic cavity. The multilobulated cystic and necrotic neoplasm was composed of interlacing streams and fascicles of neoplastic spindle cells arranged in Antoni A, and less commonly, Antoni B patterns. Verocay bodies were present also. The neoplasm was encapsulated mostly, and histomorphologic features were benign. Immunohistochemistry indicated that neoplastic cells were positive for vimentin, S100, glial fibrillary acidic protein, and nerve growth factor receptor. Reticulin histochemical staining and immunohistochemical stains for collagen IV and laminin showed a prominent basal lamina surrounding the neoplastic cells. The histologic features and results of the immunohistochemical stains confirmed peripheral nerve origin and were consistent with schwannoma. To our knowledge, this is the first case of thoracic schwannoma in a rhesus macaque and the second reported case of schwannoma in a nonhuman primate.
Collapse
Affiliation(s)
- Derron A Alves
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, USA.
| | | | | | | | | |
Collapse
|
36
|
Prabhakar S, Brenner GJ, Sung B, Messerli SM, Mao J, Sena-Esteves M, Stemmer-Rachamimov A, Tannous B, Breakefield XO. Imaging and therapy of experimental schwannomas using HSV amplicon vector-encoding apoptotic protein under Schwann cell promoter. Cancer Gene Ther 2010; 17:266-74. [PMID: 19834516 PMCID: PMC2857743 DOI: 10.1038/cgt.2009.71] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2009] [Revised: 07/04/2009] [Accepted: 09/04/2009] [Indexed: 12/27/2022]
Abstract
Schwannomas are benign tumors forming along peripheral nerves that can cause deafness, pain and paralysis. Current treatment involves surgical resection, which can damage associated nerves. To achieve tumor regression without damage to nerve fibers, we generated an HSV amplicon vector in which the apoptosis-inducing enzyme, caspase-1 (ICE), was placed under the Schwann cell-specific P0 promoter. Infection of schwannoma, neuroblastoma and fibroblastic cells in culture with ICE under the P0 promoter showed selective toxicity to schwannoma cells, while ICE under a constitutive promoter was toxic to all cell types. After direct intratumoral injection of the P0-ICE amplicon vector, we achieved marked regression of schwannoma tumors in an experimental xenograft mouse model. Injection of this amplicon vector into the sciatic nerve produced no apparent injury to the associated dorsal root ganglia neurons or myelinated nerve fibers. The P0-ICE amplicon vector provides a potential means of 'knifeless resection' of schwannoma tumors by injection of the vector into the tumor with low risk of damage to associated nerve fibers.
Collapse
Affiliation(s)
- S Prabhakar
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - GJ Brenner
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Anaesthesiology, Massachusetts General Hospital, Boston, MA, USA
| | - B Sung
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesia and Critical Care, Pain Research Group, Massachusetts General Hospital, Boston, MA, USA
| | - SM Messerli
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - J Mao
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Anesthesia and Critical Care, Pain Research Group, Massachusetts General Hospital, Boston, MA, USA
| | - M Sena-Esteves
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Program in Neuroscience, Harvard Medical School, Boston, MA, USA
| | - A Stemmer-Rachamimov
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - B Tannous
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - XO Breakefield
- Neuroscience Center, Massachusetts General Hospital, Boston, MA, USA
- Department of Neurology, Massachusetts General Hospital, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Center for Molecular Imaging Research, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
37
|
PTEN dosage is essential for neurofibroma development and malignant transformation. Proc Natl Acad Sci U S A 2009; 106:19479-84. [PMID: 19846776 DOI: 10.1073/pnas.0910398106] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Patients with neurofibromatosis type 1 (NF1) carry approximately a 10% lifetime risk of developing a malignant peripheral nerve sheath tumor (MPNST). Although the molecular mechanisms underlying NF1 to MPNST malignant transformation remain unclear, alterations of both the RAS/RAF/MAPK and PI3K/AKT/mTOR signaling pathways have been implicated. In a series of genetically engineered murine models, we perturbed RAS/RAF/MAPK or/and PTEN/PI3K/AKT pathway, individually or simultaneously, via conditional activation of K-ras oncogene or deletion of Nf1 or Pten tumor suppressor genes. Only K-Ras activation in combination with a single Pten allele deletion led to 100% penetrable development of NF lesions and subsequent progression to MPNST. Importantly, loss or decrease in PTEN expression was found in all murine MPNSTs and a majority of human NF1-associated MPNST lesions, suggesting that PTEN dosage and its controlled signaling pathways are critical for transformation of NFs to MPNST. Using noninvasive in vivo PET-CT imaging, we demonstrated that FDG can be used to identify the malignant transformation in both murine and human MPNSTs. Our data suggest that combined inhibition of RAS/RAF/MAPK and PTEN/PI3K/AKT pathways may be beneficial for patients with MPNST.
Collapse
|
38
|
Tissue-specific ablation of Prkar1a causes schwannomas by suppressing neurofibromatosis protein production. Neoplasia 2008; 10:1213-21. [PMID: 18953430 DOI: 10.1593/neo.08652] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2008] [Revised: 07/23/2008] [Accepted: 07/29/2008] [Indexed: 11/18/2022] Open
Abstract
Signaling events leading to Schwann cell tumor initiation have been extensively characterized in the context of neurofibromatosis (NF). Similar tumors are also observed in patients with the endocrine neoplasia syndrome Carney complex, which results from inactivating mutations in PRKAR1A. Loss of PRKAR1A causes enhanced protein kinase A activity, although the pathways leading to tumorigenesis are not well characterized. Tissue-specific ablation of Prkar1a in neural crest precursor cells (TEC3KO mice) causes schwannomas with nearly 80% penetrance by 10 months. These heterogeneous neoplasms were clinically characterized as genetically engineered mouse schwannomas, grades II and III. At the molecular level, analysis of the tumors revealed almost complete loss of both NF proteins, despite the fact that transcript levels were increased, implying posttranscriptional regulation. Although Erk and Akt signaling are typically enhanced in NF-associated tumors, we observed no activation of either of these pathways in TEC3KO tumors. Furthermore, the small G proteins Ras, Rac1, and RhoA are all known to be involved with NF signaling. In TEC3KO tumors, all three molecules showed modest increases in total protein, but only Rac1 showed significant activation. These data suggest that dysregulated protein kinase A activation causes tumorigenesis through pathways that overlap but are distinct from those described in NF tumorigenesis.
Collapse
|
39
|
Clark JJ, Provenzano M, Diggelmann HR, Xu N, Hansen SS, Hansen MR. The ErbB inhibitors trastuzumab and erlotinib inhibit growth of vestibular schwannoma xenografts in nude mice: a preliminary study. Otol Neurotol 2008; 29:846-53. [PMID: 18636037 PMCID: PMC2652856 DOI: 10.1097/mao.0b013e31817f7398] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To analyze the ability of ErbB inhibitors to reduce the growth of vestibular schwannoma (VS) xenografts. METHODS Vestibular schwannoma xenografts were established in the interscapular fat pad in nude mice for 4 weeks. Initially, a small cohort of animals was treated with the ErbB2 inhibitor trastuzumab or saline for 2 weeks. Animals also received bromodeoxyuridine injections to label proliferating cells. In a longer-term experiment, animals were randomized to receive trastuzumab, erlotinib (an ErbB kinase inhibitor), or placebo for 12 weeks. Tumor growth was monitored by magnetic resonance imaging during the treatment period. Cell death was analyzed by terminal deoxynucleotidyl transferase-mediated dUTP-biotin end labeling of fragmented DNA. RESULTS Tumors can be distinguished with T2-weighted magnetic resonance imaging sequences. Trastuzumab significantly reduced the proliferation of VS cells compared with control (p < 0.01) as analyzed by bromodeoxyuridine uptake. Control tumors demonstrated slight growth during the 12-week treatment period. Both trastuzumab and erlotinib significantly reduced the growth of VS xenografts (p < 0.05). Erlotinib, but not trastuzumab, resulted in a significant increase in the percentage of terminal deoxynucleotidyl transferase-mediated dUTP-biotin end labeling of fragmented DNA-positive VS cells (p < 0.01). CONCLUSION In this preliminary study, the ErbB inhibitors trastuzumab and erlotinib decreased growth of VS xenografts in nude mice, raising the possibility of using ErbB inhibitors in the management of patients with schwannomas, particularly those with neurofibromatosis Type 2.
Collapse
Affiliation(s)
- J Jason Clark
- Department of Otolaryngology-Head and Neck Surgery, University of Iowa, Iowa City, Iowa 52242-1078, USA
| | | | | | | | | | | |
Collapse
|
40
|
Roth TM, Ramamurthy P, Muir D, Wallace MR, Zhu Y, Chang L, Barald KF. Influence of hormones and hormone metabolites on the growth of Schwann cells derived from embryonic stem cells and on tumor cell lines expressing variable levels of neurofibromin. Dev Dyn 2008; 237:513-24. [PMID: 18213578 DOI: 10.1002/dvdy.21430] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Loss of neurofibromin, the protein product of the tumor suppressor gene neurofibromatosis type 1 (NF1), is associated with neurofibromas, composed largely of Schwann cells. The number and size of neurofibromas in NF1 patients have been shown to increase during pregnancy. A mouse embryonic stem cell (mESC) model was used, in which mESCs with varying levels of neurofibromin were differentiated into Schwann-like cells. NF1 cell lines derived from a malignant and a benign human tumor were used to study proliferation in response to hormones. Estrogen and androgen receptors were not expressed or expressed at very low levels in the NF1+/+ cells, at low levels in NF1+/-cells, and robust levels in NF1-/-cells. A 17beta-estradiol (E2) metabolite, 2-methoxy estradiol (2ME2) is cytotoxic to the NF1-/- malignant tumor cell line, and inhibits proliferation in the other cell lines. 2ME2 or its derivatives could provide new treatment avenues for NF1 hormone-sensitive tumors at times of greatest hormonal influence.
Collapse
Affiliation(s)
- Therese M Roth
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, Michigan 48109-2200, USA
| | | | | | | | | | | | | |
Collapse
|
41
|
James MF, Lelke JM, Maccollin M, Plotkin SR, Stemmer-Rachamimov AO, Ramesh V, Gusella JF. Modeling NF2 with human arachnoidal and meningioma cell culture systems: NF2 silencing reflects the benign character of tumor growth. Neurobiol Dis 2008; 29:278-92. [PMID: 17962031 PMCID: PMC2266821 DOI: 10.1016/j.nbd.2007.09.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2007] [Revised: 08/28/2007] [Accepted: 09/03/2007] [Indexed: 11/28/2022] Open
Abstract
Meningiomas, common tumors arising from arachnoidal cells of the meninges, may occur sporadically, or in association with the inherited disorder, neurofibromatosis 2 (NF2). Most sporadic meningiomas result from NF2 inactivation, resulting in loss of tumor suppressor merlin, implicated in regulating membrane-cytoskeletal organization. To investigate merlin function in an authentic target cell type for NF2 tumor formation, we established primary cultures from genetically-matched meningioma and normal arachnoidal tissues. Our studies revealed novel and distinct cell biological and biochemical properties unique to merlin-deficient meningioma cells compared to merlin-expressing arachnoidal and meningioma cells, and other NF2-deficient cell types. Merlin-deficient meningioma cells displayed cytoskeletal and cell contact defects, altered cell morphology and growth properties, most notably cell senescence, implicating the activation of senescence pathways in limiting benign meningioma growth. Merlin suppression by RNAi in arachnoidal cells replicated merlin-deficient meningioma features, thus establishing these cell systems as disease-relevant models for studying NF2 tumorigenesis.
Collapse
Affiliation(s)
- Marianne F James
- Molecular Neurogenetics Unit, Center for Human Genetic Research, Massachusetts General Hospital, Richard B. Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | | | | | | | | | | | | |
Collapse
|
42
|
Shamji MF, Benoit BG. Syndromic and sporadic pediatric optic pathway gliomas: review of clinical and histopathological differences and treatment implications. Neurosurg Focus 2007. [DOI: 10.3171/foc-07/11/e3] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
✓Optic pathway gliomas (OPGs) are the most common primary neoplasm of the optic pathway. These lesions usually present in childhood and can arise anywhere along the optic pathway; they occur more frequently in women; and they rarely undergo late progression. Management strategies after the initial diagnosis are controversial, compounded by the different behaviors exhibited by sporadic and syndromic tumors. Neurofibromatosis Type 1 (NF1), with aberrant oncogenic signaling and consequent predisposition to intracranial tumors, is the most common associated syndrome, with nearly 20% of NF1 patients developing OPGs. A comorbid NF1 diagnosis has implications for tumor location with greater predilection for optic nerve involvement, whereas chiasmal and postchiasmal lesions are more frequently seen in sporadic cases. Syndromic OPGs often exhibit more indolent behavior and lower rates of clinical progression, and the majority of these are diagnosed by routine neuroophthalmological screening. When treatment is indicated, however, the molecular abnormalities that constitute this syndrome can limit the available chemotherapy and radiotherapy options because clinicians fear secondary malignancy and cerebrovascular complications. Furthermore, radiotherapy early in life can impair an individual's intellectual development, endocrine function, and physical growth, thereby limiting the role of this modality in the treatment of this childhood lesion. Differential gene expression and histogenesis among sporadic and syndromic OPGs may account for the different tumor behaviors, but studies correlating specific genetic and proteomic changes with patient outcome are pending. Loss of heterozygosity at 10 and 17q are more common among patients with NF1, and Ki67 labeling intensity of 2–3% and low p53 labeling intensity seem prognostic of aggressive tumor behavior. Recent advances in the development of a preclinical mouse model of NF1-associated OPG will permit investigation into improved detection strategies and chemotherapeutic and radiotherapy treatment protocols.
Collapse
Affiliation(s)
- Mohammed F. Shamji
- 1Division of Neurosurgery, The Ottawa Hospital, Ottawa, Canada; and
- 2Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Brien G. Benoit
- 1Division of Neurosurgery, The Ottawa Hospital, Ottawa, Canada; and
| |
Collapse
|
43
|
Balasubramaniam A, Shannon P, Hodaie M, Laperriere N, Michaels H, Guha A. Glioblastoma multiforme after stereotactic radiotherapy for acoustic neuroma: case report and review of the literature. Neuro Oncol 2007; 9:447-53. [PMID: 17704364 PMCID: PMC1994102 DOI: 10.1215/15228517-2007-027] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Indications for the use of radiotherapy in the management of a variety of benign intracranial neoplastic and nonneoplastic pathologies are increasing. Although the short-term risks are minimal, the long-term risks of radiation-induced de novo secondary neoplasms or malignant progression of the primary benign tumor need to be considered. There are currently 19 reported cases of tumors linked with stereotactic radiotherapy/radiosurgery, to which we add our second institutional experience of a patient who succumbed to a glioblastoma multiforme (GBM) after stereotactic radiotherapy for an acoustic neuroma (AN). Review of these 20 cases revealed 10 de novo secondary tumors, of which eight were malignant, with six being malignant gliomas. The majority of the cases (14 of 20) involved AN, with most being in patients with neurofibromatosis-2 (NF2; 8 of 14), reflecting the large numbers and long-term use of radiotherapy for AN. Accelerated growth of the primary benign AN, some 2 to 6 years after focused radiotherapy, was found in six of eight NF2 patients, with pathological verification of a malignant nerve sheath tumor documented in most. The exact carcinogenic risk after radiotherapy is unknown but likely extremely low. However, the risk is not zero and requires discussion with the patient, with specific consideration in young patients and those with a cancer predisposition.
Collapse
Affiliation(s)
| | | | | | | | | | - Abhijit Guha
- Address correspondence to Abhijit Guha, Arthur and Sonia Labatt Brain Tumor Center, Hospital for Sick Children, University of Toronto, 399 Bathurst St., 4W-446, Toronto, ON M5T-2S8, Canada (
)
| |
Collapse
|
44
|
Cargioli TG, Ugur HC, Ramakrishna N, Chan J, Black PM, Carroll RS. Establishment of an in vivo meningioma model with human telomerase reverse transcriptase. Neurosurgery 2007; 60:750-9; discussion 759-60. [PMID: 17415213 DOI: 10.1227/01.neu.0000255397.00410.8f] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE The lack of meningioma models has hindered research on the pathogenesis and treatment of this commonly diagnosed primary brain tumor. Animal models of meningioma have been difficult to develop, especially those derived from Grade I tumors, which display very slow growth rates, senesce at early passages, and infrequently survive as explants in vivo. In this study, the authors report the establishment of two benign immortalized meningioma cell lines, Me10T and Me3TSC, that can serve as useful models of human meningioma. METHODS Tissue specimens obtained at the time of surgery were cultured in vitro and transduced with human telomerase reverse transcriptase/SV40 large T antigen to establish long-term cell lines. The telomeric activity, growth kinetics, immunophenotype, and karyotyping of the cell lines were investigated. The growth inhibitory effects of the antitumor therapies, hydroxyurea and sodium butyrate, on these cell lines were determined. In addition, immortalized cell lines were implanted subdurally into mice to confirm their ability to form tumors. RESULTS Two immortalized benign meningioma cell lines, Me10T and Me3TSC, transduced with catalytic subunit human telomerase reverse transcriptase alone or human telomerase reverse transcriptase and SV40 large T antigen, were established. The meningeal phenotype of the established cell cultures and orthotopic xenografts was confirmed by immunostaining. After subdural injection into athymic nude mice, both cell lines formed identifiable tumors with histological features and immunostaining patterns of human meningioma. CONCLUSION The Me3TSC and Me10T cell lines can serve as useful model systems for biological studies and the evaluation of novel therapies on meningioma.
Collapse
Affiliation(s)
- Theresa G Cargioli
- Department of Neurosurgery, Brigham and Women's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
45
|
Prabhakar S, Messerli SM, Stemmer-Rachamimov AO, Liu TC, Rabkin S, Martuza R, Breakefield XO. Treatment of Implantable NF2 Schwannoma Tumor Models with Oncolytic Herpes Simplex Virus G47Δ. Cancer Gene Ther 2007; 14:460-7. [PMID: 17304235 DOI: 10.1038/sj.cgt.7701037] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Schwannomas are benign tumors composed of dedifferentiated Schwann cells that form along peripheral nerves causing nerve compression often associated with pain and loss of function. Current surgical therapy involves total or subtotal surgical removal of the tumor, which may cause permanent nerve damage. In the present study, we explore an alternate means of therapy in which schwannomas are injected with a replication-conditional herpes simplex virus (HSV) vector to shrink the tumor through cell lysis during virus propagation. The oncolytic vector used, G47Delta, has deletions in HSV genes, which allow it to replicate selectively in dividing cells, sparing neurons. Two schwannoma cell lines were used to generate subcutaneous tumors in nude mice: HEI193, an immortalized human line previously established from an NF2 patient and NF2S-1, a newly generated spontaneous mouse line. Subcutaneous HEI193 tumors grew about ten times as fast as NF2S-1 tumors, and both regressed substantially following injection of G47Delta. Complete regression of HEI193 tumors was achieved in most animals, whereas all NF2S-1 tumors resumed growth within 2 weeks after vector injection. These studies provide a new schwannoma model for testing therapeutic strategies and demonstrate that oncolytic HSV vectors can be successfully used to shrink growing schwannomas.
Collapse
Affiliation(s)
- S Prabhakar
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital, Charlestown, MA 02129, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Perrin GQ, Fishbein L, Thomson SA, Thomas SL, Stephens K, Garbern JY, DeVries GH, Yachnis AT, Wallace MR, Muir D. Plexiform-like neurofibromas develop in the mouse by intraneural xenograft of an NF1 tumor-derived Schwann cell line. J Neurosci Res 2007; 85:1347-57. [PMID: 17335073 DOI: 10.1002/jnr.21226] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Plexiform neurofibromas are peripheral nerve sheath tumors that arise frequently in neurofibromatosis type 1 (NF1) and have a risk of malignant progression. Past efforts to establish xenograft models for neurofibroma involved the implantation of tumor fragments or heterogeneous primary cultures, which rarely achieved significant tumor growth. We report a practical and reproducible animal model of plexiform-like neurofibroma by xenograft of an immortal human NF1 tumor-derived Schwann cell line into the peripheral nerve of scid mice. The S100 and p75 positive sNF94.3 cell line was shown to possess a normal karyotype and have apparent full-length neurofibromin by Western blot. These cells were shown to have a constitutional NF1 microdeletion and elevated Ras-GTP activity, however, suggesting loss of normal neurofibromin function. Localized intraneural injection of the cell line sNF94.3 produced consistent and slow growing tumors that infiltrated and disrupted the host nerve. The xenograft tumors resembled plexiform neurofibromas with a low rate of proliferation, abundant extracellular matrix (hypocellularity), basal laminae, high vascularity, and mast cell infiltration. The histologic features of the developed tumors were particularly consistent with those of human plexiform neurofibroma as well. Intraneural xenograft of sNF94.3 cells enables the precise initiation of intraneural, plexiform-like tumors and provides a highly reproducible model for the study of plexiform neurofibroma tumorigenesis. This model facilitates testing of potential therapeutic interventions, including angiogenesis inhibitors, in a relevant cellular environment.
Collapse
Affiliation(s)
- George Q Perrin
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610-0244, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Riemenschneider MJ, Perry A, Reifenberger G. Histological classification and molecular genetics of meningiomas. Lancet Neurol 2006; 5:1045-54. [PMID: 17110285 DOI: 10.1016/s1474-4422(06)70625-1] [Citation(s) in RCA: 343] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Meningiomas account for up to 30% of all primary intracranial tumours. They are histologically classified according to the World Health Organization (WHO) classification of tumours of the nervous system. Most meningiomas are benign lesions of WHO grade I, whereas some meningioma variants correspond with WHO grades II and III and are associated with a higher risk of recurrence and shorter survival times. Mutations in the NF2 gene and loss of chromosome 22q are the most common genetic alterations associated with the initiation of meningiomas. With increase in tumour grade, additional progression-associated molecular aberrations can be found; however, most of the relevant genes are yet to be identified. High-throughput techniques of global genome and transcriptome analyses and new meningioma models provide increasing insight into meningioma biology and will help to identify common pathogenic pathways that may be targeted by new therapeutic approaches.
Collapse
|
48
|
Hoffman A, Blocker T, Dubielzig R, Ehrhart EJ. Feline periocular peripheral nerve sheath tumor: a case series. Vet Ophthalmol 2005; 8:153-8. [PMID: 15910367 DOI: 10.1111/j.1463-5224.2005.00341.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The objective of this retrospective study was to describe signalment, clinical presentation, surgical treatment, outcome, histopathologic, and immunohistochemical characteristics of periocular peripheral nerve sheath tumor in a series of cats. Six cats with a histologic diagnosis of peripheral nerve sheath tumor of the eyelid and/or conjunctiva were identified from 3997 feline ocular histopathologic submissions to the Comparative Ocular Pathology Laboratory of Wisconsin between 1976 and 2002. Medical records were reviewed and data were collected for signalment, eye affected, location, duration, and history of tumor recurrence. Ancillary diagnostic tests, surgical approach, and histopathology were also described. Special stains and immunohistochemistry panels were performed on the tissue. Follow-up information was obtained through telecommunications with the owner and local veterinarian. The mean age of the cats was 8 years (range: 5-15 years). There was an equal distribution of males and females. Breeds represented were two Domestic Long-haired (DLH) cats, three Domestic Short-haired (DSH) cats, and one Manx. The right eye was affected in four cats and the left eye in two cats. The superior eyelid was affected in four cats, the superior conjunctiva in two cats, and the inferior eyelid in one cat. All of the cats that underwent surgical excision, cryoablation, or laser ablation had an average of three recurrences of the tumor. Four of six cats undergoing wide excision with enucleation, exenteration or rhomboid flap had no recurrence of the tumor. Histomorphologically, all of the tumors were of spindle cell type with five of six tumors showing histologic features of Antoni type B tissue. All six tumors stained positive for vimentin. Five of six tumors stained positive for S-100 and laminin. All six tumors were negative for smooth muscle actin. Definitive histologic diagnoses of spindle cell tumors can be difficult. The presence of Antoni A and B histologic patterns as well as immunohistochemical reactivity for vimentin, S-100, and laminin suggest a diagnosis of peripheral nerve sheath tumor. Our findings suggest that local aggressive recurrence is common and that wide surgical excision combined with enucleation or exenteration may be indicated.
Collapse
|
49
|
Affiliation(s)
- Ho-Keung Ng
- Department of Pathology, The Chinese University of Hong Kong, Shatin New Territories, Hong Kong SAR, China.
| |
Collapse
|
50
|
Coming of Age in the Life of Neoplasia. Neoplasia 2004. [DOI: 10.1593/neo.6-6ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|