1
|
Gracia F, Sanchez-Laorden B, Gomez-Sanchez JA. Schwann cells in regeneration and cancer: an epithelial-mesenchymal transition perspective. Open Biol 2025; 15:240337. [PMID: 40037534 DOI: 10.1098/rsob.240337] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/13/2025] [Accepted: 02/09/2025] [Indexed: 03/06/2025] Open
Abstract
In the peripheral nervous system, glial cells, known as Schwann cells (SCs), are responsible for supporting and maintaining nerves. One of the most important characteristics of SCs is their remarkable plasticity. In various injury contexts, SCs undergo a reprogramming process that generates specialized cells to promote tissue regeneration and repair. However, in pathological conditions, this same plasticity and regenerative potential can be hijacked. Different studies highlight the activation of the epithelial-mesenchymal transition (EMT) as a driver of SC phenotypic plasticity. Although SCs are not epithelial, their neural crest origin makes EMT activation crucial for their ability to adopt repair phenotypes, mirroring the plasticity observed during development. These adaptive processes are essential for regeneration. However, EMT activation in SCs-derived tumours enhances cancer progression and aggressiveness. Furthermore, in the tumour microenvironment (TME), SCs also acquire activated phenotypes that contribute to tumour migration and invasion by activating EMT in cancer cells. In this review, we will discuss how EMT impacts SC plasticity and function from development and tissue regeneration to pathological conditions, such as cancer.
Collapse
Affiliation(s)
- Francisco Gracia
- Instituto de Neurociencias CSIC-UMH, San Juan de Alicante, 03550, Spain
| | | | - Jose A Gomez-Sanchez
- Instituto de Neurociencias CSIC-UMH, San Juan de Alicante, 03550, Spain
- Instituto de Investigacion Sanitaria y Biomedica de Alicante (ISABIAL), Alicante 03010, Spain
| |
Collapse
|
2
|
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
|
3
|
Fareez F, Wang BH, Brain I, Lu JQ. Lymphomas in patients with neurofibromatosis type 1 (NF1): another malignancy in the NF1 syndrome? Pathology 2023; 55:302-314. [PMID: 36774237 DOI: 10.1016/j.pathol.2023.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 12/22/2022] [Accepted: 01/06/2023] [Indexed: 01/21/2023]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant multisystem syndrome caused by mutations in the neurofibromin 1 (NF1) gene that encodes for the protein neurofibromin acting as a tumour suppressor. Neurofibromin functions primarily as a GTPase-activating protein for the Ras family of oncogenes, which activates many signalling pathways for cell proliferation and differentiation; without neurofibromin, Ras is constitutively activated, thereby turning on many downstream signalling pathways related to oncogenesis. Patients with NF1 have a well known predisposition for certain types of malignancies including malignant peripheral nerve sheath tumours, gliomas, and breast cancers, as well as a potential association of NF1 with lymphoproliferative disorders such as lymphomas. In this article, we review the pathophysiology and tumourigenesis of NF1, previously reported cases of cutaneous lymphomas in NF1 patients along with our case demonstration of a NF1-associated scalp B-cell lymphoma, and NF1-associated extra cutaneous lymphomas. The diagnosis of lymphomas particularly cutaneous lymphomas may be difficult in NF1 patients as they often have skin lesions and/or cutaneous/subcutaneous nodules or tumours like neurofibromas, which raises the possibility of underdiagnosed cutaneous lymphomas in NF1 patients. We also comprehensively discuss the association between NF1 and lymphomas. In summary, most studies support a potential association between NF1 and lymphomas. Further investigation is needed to clarify the association between NF1 and lymphomas in order to bring clinical awareness of possibly underdiagnosed NF1-associated lymphomas and individualised management of NF1 patients to practice.
Collapse
Affiliation(s)
- Faiha Fareez
- Department of Pathology and Molecular Medicine, Hamilton, Ontario, Canada
| | - Bill H Wang
- Department of Surgery/Neurosurgery, McMaster University, Hamilton, Ontario, Canada
| | - Ian Brain
- Department of Laboratory Medicine and Pathobiology/Hematopathology, University of Toronto, Toronto, Ontario, Canada
| | - Jian-Qiang Lu
- Department of Pathology and Molecular Medicine, Hamilton, Ontario, Canada; Department of Pathology and Molecular Medicine/Neuropathology, Hamilton General Hospital, Hamilton, Ontario, Canada.
| |
Collapse
|
4
|
Kershner LJ, Choi K, Wu J, Zhang X, Perrino M, Salomonis N, Shern JF, Ratner N. Multiple Nf1 Schwann cell populations reprogram the plexiform neurofibroma tumor microenvironment. JCI Insight 2022; 7:e154513. [PMID: 36134665 PMCID: PMC9675562 DOI: 10.1172/jci.insight.154513] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
To define alterations early in tumor formation, we studied nerve tumors in neurofibromatosis 1 (NF1), a tumor predisposition syndrome. Affected individuals develop neurofibromas, benign tumors driven by NF1 loss in Schwann cells (SCs). By comparing normal nerve cells to plexiform neurofibroma (PN) cells using single-cell and bulk RNA sequencing, we identified changes in 5 SC populations, including a de novo SC progenitor-like (SCP-like) population. Long after Nf1 loss, SC populations developed PN-specific expression of Dcn, Postn, and Cd74, with sustained expression of the injury response gene Postn and showed dramatic expansion of immune and stromal cell populations; in corresponding human PNs, the immune and stromal cells comprised 90% of cells. Comparisons between injury-related and tumor monocytes/macrophages support early monocyte recruitment and aberrant macrophage differentiation. Cross-species analysis verified each SC population and unique conserved patterns of predicted cell-cell communication in each SC population. This analysis identified PROS1-AXL, FGF-FGFR, and MIF-CD74 and its effector pathway NF-κB as deregulated in NF1 SC populations, including SCP-like cells predicted to influence other types of SCs, stromal cells, and/or immune cells in mouse and human. These findings highlight remarkable changes in multiple types of SCs and identify therapeutic targets for PN.
Collapse
Affiliation(s)
- Leah J. Kershner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kwangmin Choi
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Xiyuan Zhang
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Melissa Perrino
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| | - Nathan Salomonis
- Division of Biomedical Informatics, and
- Departments of Pediatrics and Bioinformatics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Jack F. Shern
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio, USA
| |
Collapse
|
5
|
Ge LL, Xing MY, Zhang HB, Wang ZC. Neurofibroma Development in Neurofibromatosis Type 1: Insights from Cellular Origin and Schwann Cell Lineage Development. Cancers (Basel) 2022; 14:cancers14184513. [PMID: 36139671 PMCID: PMC9497298 DOI: 10.3390/cancers14184513] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1), a genetic tumor predisposition syndrome that affects about 1 in 3000 newborns, is caused by mutations in the NF1 gene and subsequent inactivation of its encoded neurofibromin. Neurofibromin is a tumor suppressor protein involved in the downregulation of Ras signaling. Despite a diverse clinical spectrum, one of several hallmarks of NF1 is a peripheral nerve sheath tumor (PNST), which comprises mixed nervous and fibrous components. The distinct spatiotemporal characteristics of plexiform and cutaneous neurofibromas have prompted hypotheses about the origin and developmental features of these tumors, involving various cellular transition processes. METHODS We retrieved published literature from PubMed, EMBASE, and Web of Science up to 21 June 2022 and searched references cited in the selected studies to identify other relevant papers. Original articles reporting the pathogenesis of PNSTs during development were included in this review. We highlighted the Schwann cell (SC) lineage shift to better present the evolution of its corresponding cellular origin hypothesis and its important effects on the progression and malignant transformation of neurofibromas. CONCLUSIONS In this review, we summarized the vast array of evidence obtained on the full range of neurofibroma development based on cellular and molecular pathogenesis. By integrating findings relating to tumor formation, growth, and malignancy, we hope to reveal the role of SC lineage shift as well as the combined impact of additional determinants in the natural history of PNSTs.
Collapse
Affiliation(s)
- Ling-Ling Ge
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People′s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Ming-Yan Xing
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200011, China
| | - Hai-Bing Zhang
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai 200011, China
- Correspondence: (H.-B.Z.); or (Z.-C.W.); Tel.: +86-021-54920988 (H.-B.Z.); +86-021-53315120 (Z.-C.W.)
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People′s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
- Correspondence: (H.-B.Z.); or (Z.-C.W.); Tel.: +86-021-54920988 (H.-B.Z.); +86-021-53315120 (Z.-C.W.)
| |
Collapse
|
6
|
Patritti Cram J, Wu J, Coover RA, Rizvi TA, Chaney KE, Ravindran R, Cancelas JA, Spinner RJ, Ratner N. P2RY14 cAMP signaling regulates Schwann cell precursor self-renewal, proliferation, and nerve tumor initiation in a mouse model of neurofibromatosis. eLife 2022; 11:73511. [PMID: 35311647 PMCID: PMC8959601 DOI: 10.7554/elife.73511] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 01/19/2022] [Indexed: 01/05/2023] Open
Abstract
Neurofibromatosis type 1 (NF1) is characterized by nerve tumors called neurofibromas, in which Schwann cells (SCs) show deregulated RAS signaling. NF1 is also implicated in regulation of cAMP. We identified the G-protein-coupled receptor (GPCR) P2ry14 in human neurofibromas, neurofibroma-derived SC precursors (SCPs), mature SCs, and mouse SCPs. Mouse Nf1-/- SCP self-renewal was reduced by genetic or pharmacological inhibition of P2ry14. In a mouse model of NF1, genetic deletion of P2ry14 rescued low cAMP signaling, increased mouse survival, delayed neurofibroma initiation, and improved SC Remak bundles. P2ry14 signals via Gi to increase intracellular cAMP, implicating P2ry14 as a key upstream regulator of cAMP. We found that elevation of cAMP by either blocking the degradation of cAMP or by using a P2ry14 inhibitor diminished NF1-/- SCP self-renewal in vitro and neurofibroma SC proliferation in in vivo. These studies identify P2ry14 as a critical regulator of SCP self-renewal, SC proliferation, and neurofibroma initiation.
Collapse
Affiliation(s)
- Jennifer Patritti Cram
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,Neuroscience Graduate Program, University of Cincinnati College of Medicine, Cincinnati, United States
| | - Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, United States
| | - Robert A Coover
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Tilat A Rizvi
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Katherine E Chaney
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States
| | - Ramya Ravindran
- Molecular and Developmental Biology, Cincinnati Children's Hospital, Cincinnati, United States
| | - Jose A Cancelas
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,Hoxworth Blood Center, College of Medicine, University of Cincinnati, Cincinnati, United States
| | - Robert J Spinner
- Department of Neurosurgery, Mayo Clinic, Rochester, United States
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cancer & Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, United States
| |
Collapse
|
7
|
Preis E, Schulze J, Gutberlet B, Pinnapireddy SR, Jedelská J, Bakowsky U. The chorioallantoic membrane as a bio-barrier model for the evaluation of nanoscale drug delivery systems for tumour therapy. Adv Drug Deliv Rev 2021; 174:317-336. [PMID: 33905805 DOI: 10.1016/j.addr.2021.04.022] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/29/2021] [Accepted: 04/20/2021] [Indexed: 02/06/2023]
Abstract
In 2010, the European Parliament and the European Union adopted a directive on the protection of animals used for scientific purposes. The directive aims to protect animals in scientific research, with the final goal of complete replacement of procedures on live animals for scientific and educational purposes as soon as it is scientifically viable. Furthermore, the directive announces the implementation of the 3Rs principle: "When choosing methods, the principles of replacement, reduction and refinement should be implemented through a strict hierarchy of the requirement to use alternative methods." The visibility, accessibility, and the rapid growth of the chorioallantoic membrane (CAM) offers a clear advantage for various manipulations and for the simulation of different Bio-Barriers according to the 3R principle. The extensive vascularisation on the CAM provides an excellent substrate for the cultivation of tumour cells or tumour xenografts which could be used for the therapeutic evaluation of nanoscale drug delivery systems. The tumour can be targeted either by topical application, intratumoural injection or i.v. injection. Different application sites and biological barriers can be examined within a single model.
Collapse
Affiliation(s)
- Eduard Preis
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Jan Schulze
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Bernd Gutberlet
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany
| | - Shashank Reddy Pinnapireddy
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany; CSL Behring Innovation GmbH, Emil-von-Behring-Str. 76, 35041 Marburg, Germany
| | - Jarmila Jedelská
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany; Center for Tumor Biology and Immunology, Core Facility for Small Animal MRI, Hans-Meerwein Str. 3, 35043 Marburg, Germany
| | - Udo Bakowsky
- Department of Pharmaceutics and Biopharmaceutics, University of Marburg, Robert-Koch-Str. 4, 35037 Marburg, Germany.
| |
Collapse
|
8
|
Abstract
Neurofibromatosis type I (NF1) is a debilitating inherited tumor syndrome affecting around 1 in 3000 people. Patients present with a variety of tumors caused by biallelic loss of the tumor suppressor neurofibromin (NF1), a negative regulator of Ras signaling. While the mechanism of tumor formation is similar in the majority of NF1 cases, the clinical spectrum of tumors can vary depending on spatiotemporal loss of heterozygosity of NF1 in cells derived from the neural crest during development. The hallmark lesions that give NF1 its namesake are neurofibromas, which are benign Schwann cell tumors composed of nervous and fibrous tissue. Neurofibromas can be found in the skin (cutaneous neurofibroma) or deeper in body near nerve plexuses (plexiform neurofibroma). While neurofibromas have been known to be Schwann cell tumors for many years, the exact timing and initiating cell has remained elusive. This has led to difficulties in developing animal models and successful therapies for NF1. A culmination of recent genetic studies has finally begun to shed light on the detailed cellular origins of neurofibromatosis. In this review, we will examine the hunt for neurofibroma tumor cells of origin through a historical lens, detailing the genetic systems used to delineate the source of plexiform and cutaneous neurofibromas. Through these novel findings, we can better understand the cellular, temporal, and developmental context during tumor initiation. By leveraging this data, we hope to uncover new therapeutic targets and mechanisms to treat NF1 patients.
Collapse
Affiliation(s)
- Stephen Li
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas.,Medical Scientist Training Program, University of Texas Southwestern Medical Center, Dallas.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas
| | - Zhiguo Chen
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center, Dallas.,Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas.,Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas.,Neurofibromatosis Clinic, University of Texas Southwestern Medical Center, Dallas
| |
Collapse
|
9
|
Azhar AF, Bittle JSH, Kwarcinski TJ, Hinshelwood JR. Embolization of a hemorrhaging abdominal plexiform neurofibroma. Proc (Bayl Univ Med Cent) 2020; 33:448-450. [PMID: 32675983 DOI: 10.1080/08998280.2020.1738853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 02/26/2020] [Accepted: 02/27/2020] [Indexed: 10/24/2022] Open
Abstract
In rare instances, neurofibromas may spontaneously bleed. In this case, a 21-year-old woman with a known cutaneous neurofibroma presented with a rapidly enlarging right abdominal wall mass. After embolization of a focal pseudoaneurysm and the right internal mammary artery, her hemoglobin continued to decline. The right internal mammary artery and inferior epigastric artery were then embolized. The patient eventually underwent mass resection, hematoma evacuation, and flap reconstruction. Final surgical pathology confirmed the expected diagnosis of plexiform neurofibroma. The Schwann cells of plexiform neurofibromas, present in approximately 50% of patients with neurofibromatosis-1, have invasive and angiogenic properties, often resulting in hemorrhage.
Collapse
Affiliation(s)
- Aaminah F Azhar
- College of Medicine, Texas A&M Health Sciences CenterDallasTexas
| | | | | | - Jonathan R Hinshelwood
- Department of Radiology, Prisma Health Systems/Greenville Medical CenterGreenvilleSouth Carolina
| |
Collapse
|
10
|
Schneider-Stock R, Ribatti D. The CAM Assay as an Alternative In Vivo Model for Drug Testing. Handb Exp Pharmacol 2020; 265:303-323. [PMID: 32776283 DOI: 10.1007/164_2020_375] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In the last decade, the chicken chorioallantoic membrane (CAM) assay has been re-discovered in cancer research to study the molecular mechanisms of anti-cancer drug effects. Literature about the CAM assay as an alternative in vivo cancer xenograft model according to the 3R principles has exploded in the last 3 years. Following a summary of the basic knowledge about the chicken embryo, we compare advantages and disadvantages with the classical mouse xenograft model, exemplify established and innovative imaging techniques that are used in the CAM model, and give examples of its successful utilization for studying major hallmarks of cancer such as angiogenesis, proliferation, invasion, and metastasis.
Collapse
Affiliation(s)
- Regine Schneider-Stock
- Experimental Tumorpathology, Institute of Pathology, University Hospital, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - Domenico Ribatti
- Department of Basic Medical Sciences, Neurosciences and Sensory Organs, University of Bari Medical School, Bari, Italy
| |
Collapse
|
11
|
Nehete LS, Konar SK, Nandeesh BN, Shukla D, Devi BI. Atypical presentation of extraspinal neurofibroma presenting with acute-onset monoparesis and Horner's syndrome: Case report and review of literature. JOURNAL OF CRANIOVERTEBRAL JUNCTION AND SPINE 2019; 10:188-191. [PMID: 31772433 PMCID: PMC6868537 DOI: 10.4103/jcvjs.jcvjs_86_18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The clinical presentation of spinal or extraspinal neurofibroma is radiculopathy or myelopathy, pain, and motor weakness. Extraspinal neurofibroma presenting with acute-onset monoparesis and Horner's syndrome is very rare. We report the case of a 55-year-old female who presented with acute-onset monoparesis of the left upper limb along with left-side drooping of the eyelid. Imaging revealed C6–D2 extraspinal solitary mass lesion lateral to spinous process with bleed without intraspinal component. The patient underwent an anterior cervical approach and excision of the tumor. Final biopsy report was a neurofibroma. At 3-year follow-up, she recovered from motor weakness, and Horner's syndrome subsided. Extraspinal neurofibroma can present with acute bleed, and surgical outcome is superior in early intervention.
Collapse
Affiliation(s)
- Lokesh S Nehete
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Subhas K Konar
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - B N Nandeesh
- Department of Neuropathology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Dhaval Shukla
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - B Indira Devi
- Department of Neurosurgery, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| |
Collapse
|
12
|
Jia J, Zhang H, Zhang H, Du H, Liu W, Shu M. Activated androgen receptor accelerates angiogenesis in cutaneous neurofibroma by regulating VEGFA transcription. Int J Oncol 2019; 55:157-166. [PMID: 31059067 DOI: 10.3892/ijo.2019.4797] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 04/16/2019] [Indexed: 11/05/2022] Open
Abstract
Accumulating evidence has demonstrated the significant progression of cutaneous neurofibroma (cNF) without necrosis during puberty. However, the molecular events involved in this process remain unclear. The alteration of the steroid hormone levels during puberty has led to the investigation of the expression levels of the androgen receptor (AR). A positive correlation between AR expression and microvessel density has been reported in human cNF tissues in combination with enhanced endothelial cell tube formation in vitro. In addition, activated AR signaling can promote neurofibroma cell growth in vivo and in vitro and tube formation in vitro. In the present study, AR was shown to bind directly to the promoter of vascular endothelial growth factor A (VEGFA), a key factor involved in angiogenesis, and to sequentially induce its expression. Furthermore, the AR inhibitor, MDV3100, downregulated VEGFA expression and abolished endothelial cell recruitment and tube formation. Taken collectively, the findings of this study revealed that AR signaling enhanced tumor growth and angiogenesis in cNF by regulating VEGFA transcription. However, whether AR can be regarded a therapeutic target for cNF requires further investigation.
Collapse
Affiliation(s)
- Jing Jia
- Department of Plastic, Cosmetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Haibao Zhang
- Key Laboratory for Tumor Precision Medicine of Shaanxi Province, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Hongke Zhang
- Department of Plastic, Cosmetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Huicong Du
- Department of Plastic, Cosmetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Wenbo Liu
- Department of Plastic, Cosmetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Maoguo Shu
- Department of Plastic, Cosmetic and Maxillofacial Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
13
|
Meyerholz DK, Ofori-Amanfo GK, Leidinger MR, Goeken JA, Khanna R, Sieren JC, Darbro BW, Quelle DE, Weimer JM. Immunohistochemical Markers for Prospective Studies in Neurofibromatosis-1 Porcine Models. J Histochem Cytochem 2017; 65:607-618. [PMID: 28846462 DOI: 10.1369/0022155417729357] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a common, cancer-predisposing disease caused by mutations in the NF1 tumor gene. Patients with NF1 have an increased risk for benign and malignant tumors of the nervous system (e.g., neurofibromas, malignant peripheral nerve sheath tumors, gliomas) and other tissues (e.g., leukemias, rhabdomyosarcoma, etc.) as well as increased susceptibility to learning disabilities, chronic pain/migraines, hypertension, pigmentary changes, and developmental lesions (e.g., tibial pseudoarthrosis). Pigs are an attractive and upcoming animal model for future NF1 studies, but a potential limitation to porcine model research has been the lack of validated reagents for direct translational study to humans. To address that issue, we used formalin-fixed tissues (human and pigs) to evaluate select immunohistochemical markers (activated caspase-3, allograft inflammatory factor-1, beta-tubulin III, calbindin D, CD13, CD20, desmin, epithelial membrane antigen, glial fibrillary acidic protein, glucose transporter-1, laminin, myelin basic protein, myoglobin, proliferating cell nuclear antigen, S100, vimentin, and von Willebrand factor). The markers were validated by comparing known expression and localization in human and pig tissues. Validation of these markers on fixed tissues will facilitate prospective immunohistochemical studies of NF1 pigs, as well as other pig models, in a more efficient, reproducible, and translationally relevant manner.
Collapse
Affiliation(s)
| | | | | | | | - Rajesh Khanna
- University of Iowa, Iowa City, Iowa, Departments of Pharmacology and Anesthesiology, College of Medicine, University of Arizona, Tucson, Arizona.,Departments of Pharmacology and Anesthesiology, College of Medicine, University of Arizona, Tucson, Arizona
| | | | | | - Dawn E Quelle
- Department of Pathology.,Department of Pediatrics.,Department of Pharmacology
| | - Jill M Weimer
- Pediatrics and Rare Disease Group, Sanford Research, Sioux Falls, South Dakota.,Department of Pediatrics, University of South Dakota, Vermillion, South Dakota
| |
Collapse
|
14
|
Rosenbaum T, Patrie KM, Ratner N. Neurofibromatosis Type 1: Genetic and Cellular Mechanisms of Peripheral Nerve Tumor Formation. Neuroscientist 2016. [DOI: 10.1177/107385849700300614] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Neurofibromatosis type 1 (NF1) is among the most common inherited human diseases. The NF1 protein is a Ras-GTPase activating protein, positioning NF1 in important intracellular signaling pathways. Patients with mutations in the NF1 gene can develop benign peripheral nerve tumors (neurofibromas), learning disabilities, and/or benign optic nerve gliomas, in addition to abnormalities unassociated with the nervous system. The NF1 gene is believed to act as a tumor suppressor. How NF1 mutations relate to benign features of NF1 is the subject of active investigation. Studies using transgenic mice with NF1 mutations and cells derived from these mice have yielded exciting new data, implicating multiple cell types mutant at NF1 and possibly factors in the environment in the pathogenesis of benign neurofibromas. NEUROSCIENTIST 3:412-420, 1997
Collapse
Affiliation(s)
| | - Kevin M. Patrie
- Department of Pediatrics Düsseldorf Department of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine Cincinnati, Ohio
| | - Nancy Ratner
- Department of Pediatrics Düsseldorf Department of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine Cincinnati, Ohio
| |
Collapse
|
15
|
Demestre M, Terzi MY, Mautner V, Vajkoczy P, Kurtz A, Piña AL. Effects of pigment epithelium derived factor (PEDF) on malignant peripheral nerve sheath tumours (MPNSTs). J Neurooncol 2014; 115:391-9. [PMID: 24078214 DOI: 10.1007/s11060-013-1252-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2013] [Accepted: 09/17/2013] [Indexed: 11/30/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an inherited genetic disease affecting 1 in 3,500 individuals. A prominent feature of NF1 is the formation of benign tumours of the peripheral nerve sheath (neurofibromas). However, these can become malignant and form highly metastatic malignant peripheral nerve sheath tumours (MPNST), which are usually fatal despite aggressive surgery, chemotherapy, and radiotherapy. Recent studies have shown that pigment epithelium-derived factor (PEDF) can induce differentiation and inhibit angiogenesis in several kinds of tumours. The present study was designed to determine the in vitro and in vivo effects of PEDF on MPNST angiogenesis and tumour growth. PEDF inhibited proliferation and augmented apoptosis in S462 MPNST cells after 48 h of treatment in culture. In xenografts of S462 MPNST cells in athymic nude mice, PEDF suppressed MPNST tumour burden, due mainly to inhibition of angiogenesis. These results demonstrate for the first time inhibitory effects of PEDF on the growth of human MPNST via induction of anti-angiogenesis and apoptosis. Our results suggest that PEDF could be a novel approach for future therapeutic purposes against MPNST.
Collapse
|
16
|
The characterisation of Pax3 expressant cells in adult peripheral nerve. PLoS One 2013; 8:e59184. [PMID: 23527126 PMCID: PMC3602598 DOI: 10.1371/journal.pone.0059184] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/12/2013] [Indexed: 12/25/2022] Open
Abstract
Pax3 has numerous integral functions in embryonic tissue morphogenesis and knowledge of its complex function in cells of adult tissue continues to unfold. Across a variety of adult tissue lineages, the role of Pax3 is principally linked to maintenance of the tissue’s resident stem/progenitor cell population. In adult peripheral nerves, Pax3 is reported to be expressed in nonmyelinating Schwann cells, however, little is known about the purpose of this expression. Based on the evidence of the role of Pax3 in other adult tissue stem and progenitor cells, it was hypothesised that the cells in adult peripheral nerve that express Pax3 may be peripheral glioblasts. Here, methods have been developed for identification and visualisation of Pax3 expressant cells in normal 60 day old mouse peripheral nerve that allowed morphological and phenotypic distinctions to be made between Pax3 expressing cells and other nonmyelinating Schwann cells. The distinctions described provide compelling support for a resident glioblast population in adult mouse peripheral nerve.
Collapse
|
17
|
Gong M, Ma J, Li M, Zhou M, Hock JM, Yu X. MicroRNA-204 critically regulates carcinogenesis in malignant peripheral nerve sheath tumors. Neuro Oncol 2012; 14:1007-17. [PMID: 22718995 DOI: 10.1093/neuonc/nos124] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are highly aggressive soft tissue sarcomas accounting for 3%-10% of all soft tissue sarcomas. Neurofibromatosis type 1 (NF1) is the most important known risk factor. MPNSTs are often diagnosed at an advanced stage when distant metastases have developed. Although surgical resection remains the main treatment for MPNSTs, complete surgical resection is rarely possible. The prognosis for patients with MPNSTs is poor. There is an urgent need for improved therapies. To this end, we investigated whether microRNA (miR), specifically miR-204, might be implicated in MPNSTs because it is located at a cancer-associated genomic region exhibiting high frequency of loss of heterozygosity in tumors. We show that miR-204 expression is downregulated in NF1 and non-NF1 MPNST tumor tissues and in tumor cell lines. Restoring miR-204 expression in MPNST cell lines STS26T (non-NF1), ST88-14 (NF1), and T265p21 (NF1) significantly reduces cellular proliferation, migration, and invasion in vitro. Restoring miR-204 expression in STS26T decreases tumor growth and malignant progression in vivo. We also report that miR-204 inhibits Ras signaling and expression of high mobility group gene A2. These findings support the hypothesis that miR-204 plays critical roles in MPNST development and tumor progression. miR-204 may represent a novel biomarker for diagnosis and a candidate target with which to develop effective therapies for MPNSTs.
Collapse
Affiliation(s)
- Meng Gong
- Laboratory of Endocrinology and Metabolism, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, P.R. China
| | | | | | | | | | | |
Collapse
|
18
|
Staser K, Yang FC, Clapp DW. Pathogenesis of plexiform neurofibroma: tumor-stromal/hematopoietic interactions in tumor progression. ANNUAL REVIEW OF PATHOLOGY 2011; 7:469-95. [PMID: 22077553 PMCID: PMC3694738 DOI: 10.1146/annurev-pathol-011811-132441] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a genetic disease that results from either heritable or spontaneous autosomal dominant mutations in the NF1 gene. A second-hit mutation precedes the predominant NF1 neoplasms, which include myeloid leukemia, optic glioma, and plexiform neurofibroma. Despite this requisite NF1 loss of heterozygosity in the tumor cell of origin, nontumorigenic cells contribute to both generalized and specific disease manifestations. In mouse models of plexiform neurofibroma formation, Nf1 haploinsufficient mast cells promote inflammation, accelerating tumor formation and growth. These recruited mast cells, hematopoietic effector cells long known to permeate neurofibroma tissue, mediate key mitogenic signals that contribute to vascular ingrowth, collagen deposition, and tumor growth. Thus, the plexiform neurofibroma microenvironment involves a tumor/stromal interaction with the hematopoietic system that depends, at the molecular level, on a stem cell factor/c-kit-mediated signaling axis. These observations parallel findings in other NF1 disease manifestations and are clearly relevant to medical management of these neurofibromas.
Collapse
Affiliation(s)
- Karl Staser
- Department of Biochemistry, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | |
Collapse
|
19
|
Arima Y, Hayashi H, Kamata K, Goto TM, Sasaki M, Kuramochi A, Saya H. Decreased expression of neurofibromin contributes to epithelial-mesenchymal transition in neurofibromatosis type 1. Exp Dermatol 2011; 19:e136-41. [PMID: 20002172 DOI: 10.1111/j.1600-0625.2009.01017.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Plexiform and/or dermal neurofibromas are nerve sheath tumors of the peripheral nervous system that are usually present in individuals with neurofibromatosis type 1 (NF1). Neurofibromas arise from Schwann cells with biallelic inactivation of NF1, the gene that encodes neurofibromin. This protein is responsible for regulation of the Ras-mediated pathway, which has been shown to play a crucial role in epithelial-to-mesenchymal transition (EMT). EMT is a biological process that occurs during embryogenesis and wound healing and is involved in pathological processes such as organ fibrosis and cancer metastasis. However, the relationship between neurofibromin and EMT has not been elucidated. We investigated whether the EMT-related signaling pathway was upregulated in NF1-associated neurofibromas and Schwann cells by assessing the expression levels of the EMT-related transcription factors Snail, Slug, Twist, ZEB1 and ZEB2. Immunohistochemical studies and quantitative reverse transcription polymerase chain reaction revealed an increase in the expression levels of EMT-related transcription factors in neurofibroma specimens and NF1-derived Schwann cells (sNF96.2). In addition, the silencing of NF1 by siRNA induced the expression of EMT-related transcription factors in normal human Schwann cells and in epithelial-like breast cancer cells. Our findings suggest that the loss of neurofibromin activated the EMT-related signaling pathway and that the excessive mesenchymal reaction may play a key role in the development of NF1-associated neurofibromas.
Collapse
Affiliation(s)
- Yoshimi Arima
- Division of Gene Regulation, Institute for Advanced Medical Research, School of Medicine, Keio University, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
20
|
Tai WC, Wang ST, Wu CS, Lin TY, Wu MT. Chemokine receptor CCR3 is important for migration of mast cells in neurofibroma. DERMATOL SIN 2010. [DOI: 10.1016/s1027-8117(10)60033-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
21
|
Wood MD, Sanchez Y. Deregulated Ras signaling compromises DNA damage checkpoint recovery in S. cerevisiae. Cell Cycle 2010; 9:3353-63. [PMID: 20716966 DOI: 10.4161/cc.9.16.12713] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The DNA damage checkpoint maintains genome stability by arresting the cell cycle and promoting DNA repair under genotoxic stress. Cells must downregulate the checkpoint signaling pathways in order to resume cell division after completing DNA repair. While the mechanisms of checkpoint activation have been well-characterized, the process of checkpoint recovery, and the signals regulating it, has only recently been investigated. We have identified a new role for the Ras signaling pathway as a regulator of DNA damage checkpoint recovery. Here we report that in budding yeast, deletion of the IRA1 and IRA2 genes encoding negative regulators of Ras prevents cellular recovery from a DNA damage induced arrest. The checkpoint kinase Rad53 is dephosphorylated in an IRA-deficient strain, indicating that recovery failure is not caused by constitutive checkpoint pathway activation. The ira1Δ ira2Δ recovery defect requires the checkpoint kinase Chk1 and the cAMP-dependent protein kinase (PKA) catalytic subunit Tpk2. Furthermore, PKA phosphorylation sites on the anaphase promoting complex specificity factor Cdc20 are required for the recovery defect, indicating a link between the recovery defect and PKA regulation of mitosis. This work identifies a new signaling pathway that can regulate DNA damage checkpoint recovery and implicates the Ras signaling pathway as an important regulator of mitotic events.
Collapse
Affiliation(s)
- Matthew D Wood
- Department of Pharmacology and Toxicology, Norris Cotton Cancer Center, Dartmouth Medical School, Hanover, NH, USA
| | | |
Collapse
|
22
|
Lancaster E, Elman LB, Scherer SS. A patient with neurofibromatosis type 1 and Charcot-Marie-Tooth disease type 1B. Muscle Nerve 2010; 41:555-8. [PMID: 19918771 DOI: 10.1002/mus.21546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We describe a patient with both neurofibromatosis type 1 and Charcot-Marie-Tooth disease type 1B. Although one might expect an overwhelming tumor burden due to the combination of these two disorders, the two mutations did not appear to interact.
Collapse
Affiliation(s)
- Eric Lancaster
- Department of Neurology, University of Pennsylvania Medical Center, 3400 Spruce Street, 3W Gates Neurology, Philadelphia, Pennsylvania 19104, USA.
| | | | | |
Collapse
|
23
|
Miller SJ, Jessen WJ, Mehta T, Hardiman A, Sites E, Kaiser S, Jegga AG, Li H, Upadhyaya M, Giovannini M, Muir D, Wallace MR, Lopez E, Serra E, Nielsen GP, Lazaro C, Stemmer-Rachamimov A, Page G, Aronow BJ, Ratner N. Integrative genomic analyses of neurofibromatosis tumours identify SOX9 as a biomarker and survival gene. EMBO Mol Med 2010; 1:236-48. [PMID: 20049725 PMCID: PMC3378132 DOI: 10.1002/emmm.200900027] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
Understanding the biological pathways critical for common neurofibromatosis type 1 (NF1) peripheral nerve tumours is essential, as there is a lack of tumour biomarkers, prognostic factors and therapeutics. We used gene expression profiling to define transcriptional changes between primary normal Schwann cells (n = 10), NF1-derived primary benign neurofibroma Schwann cells (NFSCs) (n = 22), malignant peripheral nerve sheath tumour (MPNST) cell lines (n = 13), benign neurofibromas (NF) (n = 26) and MPNST (n = 6). Dermal and plexiform NFs were indistinguishable. A prominent theme in the analysis was aberrant differentiation. NFs repressed gene programs normally active in Schwann cell precursors and immature Schwann cells. MPNST signatures strongly differed; genes up-regulated in sarcomas were significantly enriched for genes activated in neural crest cells. We validated the differential expression of 82 genes including the neural crest transcription factor SOX9 and SOX9 predicted targets. SOX9 immunoreactivity was robust in NF and MPSNT tissue sections and targeting SOX9 – strongly expressed in NF1-related tumours – caused MPNST cell death. SOX9 is a biomarker of NF and MPNST, and possibly a therapeutic target in NF1.
Collapse
Affiliation(s)
- Shyra J Miller
- Division of Experimental Hematology, Cincinnati Children's Hospital, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Gottfried ON, Viskochil DH, Couldwell WT. Neurofibromatosis Type 1 and tumorigenesis: molecular mechanisms and therapeutic implications. Neurosurg Focus 2010; 28:E8. [DOI: 10.3171/2009.11.focus09221] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Neurofibromatosis Type 1 (NF1) is a common autosomal dominant disease characterized by complex and multicellular neurofibroma tumors, and less frequently by malignant peripheral nerve sheath tumors (MPNSTs) and optic nerve gliomas. Significant advances have been made in elucidating the cellular, genetic, and molecular biology involved in tumor formation in NF1. Neurofibromatosis Type 1 is caused by germline mutations of the NF1 tumor suppressor gene, which generally result in decreased intracellular neurofibromin protein levels, leading to increased cascade Ras signaling to its downstream effectors. Multiple key pathways are involved with the development of tumors in NF1, including Ras/mitogen-activated protein kinase (MAPK) and Akt/mammalian target of rapamycin (mTOR). Interestingly, recent studies demonstrate that multiple other developmental syndromes (in addition to NF1) share phenotypic features resulting from germline mutations in genes responsible for components of the Ras/MAPK pathway. In general, a somatic loss of the second NF1 allele, also referred to as loss of heterozygosity, in the progenitor cell, either the Schwann cell or its precursor, combined with haploinsufficiency in multiple supporting cells is required for tumor formation. Importantly, a complex series of interactions with these other cell types in neurofibroma tumorigenesis is mediated by abnormal expression of growth factors and their receptors and modification of gene expression, a key example of which is the process of recruitment and involvement of the NF1+/– heterozygous mast cell. In general, for malignant transformation to occur, there must be accumulation of additional mutations of multiple genes including INK4A/ARF and P53, with resulting abnormalities of their respective signal cascades. Further, abnormalities of the NF1 gene and molecular cascade described above have been implicated in the tumorigenesis of NF1 and some sporadically occurring gliomas, and thus, these treatment options may have wider applicability. Finally, increased knowledge of molecular and cellular mechanisms involved with NF1 tumorigenesis has led to multiple preclinical and clinical studies of targeted therapy, including the mTOR inhibitor rapamycin, which is demonstrating promising preclinical results for treatment of MPNSTs and gliomas.
Collapse
Affiliation(s)
| | - David H. Viskochil
- 2Department of Pediatrics, Division of Genetics, University of Utah, Salt Lake City, Utah
| | | |
Collapse
|
25
|
Eckert JM, Byer SJ, Clodfelder-Miller BJ, Carroll SL. Neuregulin-1 beta and neuregulin-1 alpha differentially affect the migration and invasion of malignant peripheral nerve sheath tumor cells. Glia 2009; 57:1501-20. [PMID: 19306381 PMCID: PMC2744852 DOI: 10.1002/glia.20866] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs) are the most common malignancy associated with neurofibromatosis Type 1 (NF1). These Schwann cell lineage-derived sarcomas aggressively invade adjacent nerve and soft tissue, frequently precluding surgical resection. Little is known regarding the mechanisms underlying this invasive behavior. We have shown that MPNSTs express neuregulin-1 (NRG-1) beta isoforms, which promote Schwann cell migration during development, and NRG-1 alpha isoforms, whose effects on Schwann cells are poorly understood. Hypothesizing that NRG-1 beta and/or NRG-1 alpha promote MPNST invasion, we found that NRG-1 beta promoted MPNST migration in a substrate-specific manner, markedly enhancing migration on laminin but not on collagen type I or fibronectin. The NRG-1 receptors erbB3 and erbB4 were present in MPNST invadopodia (processes mediating invasion), partially colocalized with focal adhesion kinase and the laminin receptor beta(1)-integrin and coimmunoprecipitated with beta(1)-integrin. NRG-1 beta stimulated human and murine MPNST cell migration and invasion in a concentration-dependent manner in three-dimensional migration assays, acting as a chemotactic factor. Both baseline and NRG-1 beta-induced migration were erbB-dependent and required the action of MEK 1/2, SAPK/JNK, PI-3 kinase, Src family kinases and ROCK-I/II. In contrast, NRG-1 alpha had no effect on the migration and invasion of some MPNST lines and inhibited the migration of others. While NRG-1 beta potently and persistently activated Erk 1/2, SAPK/JNK, Akt and Src family kinases, NRG-1 alpha did not activate Akt and activated these other kinases with kinetics distinct from those evident in NRG-1 beta-stimulated cells. These findings suggest that NRG-1 beta enhances MPNST migration and that NRG-1 beta and NRG-1 alpha differentially modulate this process.
Collapse
Affiliation(s)
- Jenell M Eckert
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama 35294-0017, USA
| | | | | | | |
Collapse
|
26
|
Parrinello S, Lloyd AC. Neurofibroma development in NF1--insights into tumour initiation. Trends Cell Biol 2009; 19:395-403. [PMID: 19615906 DOI: 10.1016/j.tcb.2009.05.003] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2009] [Revised: 05/22/2009] [Accepted: 05/22/2009] [Indexed: 12/31/2022]
Abstract
Dissecting the early steps of tumourigenesis is key to our understanding of cancer biology. However, lack of knowledge of initiating mutations and the target 'cell of origin' has slowed progress towards this goal. Genetically engineered mouse models of the tumour-predisposition syndrome neurofibromatosis type-1 provide a rare opportunity to study tumour initiation resulting from a known genetic change in a known cell type. Recent exciting work using these models now sheds more light onto early tumourigenesis. Here, we discuss the studies that have identified mature differentiated Schwann cells as the cell of origin and revealed the molecular and cellular mechanisms of neurofibroma initiation. A novel dual and opposing role for the microenvironment, from pro-differentiative to pro-carcinogenic, has emerged.
Collapse
Affiliation(s)
- Simona Parrinello
- MRC Laboratory for Molecular Cell Biology and the UCL Cancer Institute, University College London, Gower Street, London WC1E 6BT, UK
| | | |
Collapse
|
27
|
Abstract
Neurofibromas are benign tumors of peripheral nerve that occur sporadically or in patients with the autosomal dominant tumor predisposition syndrome neurofibromatosis type 1 (NF1). Multiple neurofibroma subtypes exist which differ in their site of occurrence, their association with NF1, and their tendency to undergo transformation to become malignant peripheral nerve sheath tumors (MPNSTs), the most common malignancy associated with NF1. Most NF1 patients carry a constitutional mutation of the NF1 tumor suppressor gene. Neurofibromas develop in these patients when an unknown cell type in the Schwann cell lineage loses its remaining functional NF1 gene and initiates a complex series of interactions with other cell types; these interactions may be influenced by aberrant expression of growth factors and growth factor receptors and the action of modifier genes. Cells within certain neurofibroma subtypes subsequently accumulate additional mutations affecting the p19(ARF)-MDM2-TP53 and p16INK4A-Rb signaling cascades, mutations of other as yet unidentified genes, and amplification of growth factor receptor genes, resulting in their transformation into MPNSTs. These observations have been validated using a variety of transgenic and knockout mouse models that recapitulate neurofibroma and MPNST pathogenesis. A new generation of mouse models is also providing important new insights into the identity of the cell type in the Schwann cell lineage that gives rise to neurofibromas. Our improving understanding of the mechanisms underlying the pathogenesis of neurofibromas and MPNSTs raises intriguing new questions about the origin and pathogenesis of these neoplasms and establishes models for the development of new therapies targeting these neoplasms.
Collapse
Affiliation(s)
- Steven L. Carroll
- Division of Neuropathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294-0017
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Research Foundation, Cincinnati Children’s Hospital Medical Center, University of Cincinnati College of Medicine, Cincinnati, OH 45229
| |
Collapse
|
28
|
Zheng H, Chang L, Patel N, Yang J, Lowe L, Burns DK, Zhu Y. Induction of abnormal proliferation by nonmyelinating schwann cells triggers neurofibroma formation. Cancer Cell 2008; 13:117-28. [PMID: 18242512 DOI: 10.1016/j.ccr.2008.01.002] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 11/12/2007] [Accepted: 01/03/2008] [Indexed: 01/09/2023]
Abstract
Recent evidence suggests that alterations in the self-renewal program of stem/progenitor cells can cause tumorigenesis. By utilizing genetically engineered mouse models of neurofibromatosis type 1 (NF1), we demonstrated that plexiform neurofibroma, the only benign peripheral nerve sheath tumor with potential for malignant transformation, results from Nf1 deficiency in fetal stem/progenitor cells of peripheral nerves. Surprisingly, this did not cause hyperproliferation or tumorigenesis in early postnatal period. Instead, peripheral nerve development appeared largely normal in the absence of Nf1 except for abnormal Remak bundles, the nonmyelinated axon-Schwann cell unit, identified in postnatal mutant nerves. Subsequent degeneration of abnormal Remak bundles was accompanied by initial expansion of nonmyelinating Schwann cells. We suggest abnormally differentiated Remak bundles as a cell of origin for plexiform neurofibroma.
Collapse
Affiliation(s)
- Huarui Zheng
- Division of Molecular Medicine and Genetics, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Wu J, Williams JP, Rizvi TA, Kordich JJ, Witte D, Meijer D, Stemmer-Rachamimov AO, Cancelas JA, Ratner N. Plexiform and dermal neurofibromas and pigmentation are caused by Nf1 loss in desert hedgehog-expressing cells. Cancer Cell 2008; 13:105-16. [PMID: 18242511 PMCID: PMC2846699 DOI: 10.1016/j.ccr.2007.12.027] [Citation(s) in RCA: 172] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 10/12/2007] [Accepted: 12/26/2007] [Indexed: 12/14/2022]
Abstract
Neurofibromatosis type 1 (Nf1) mutation predisposes to benign peripheral nerve (glial) tumors called neurofibromas. The point(s) in development when Nf1 loss promotes neurofibroma formation are unknown. We show that inactivation of Nf1 in the glial lineage in vitro at embryonic day 12.5 + 1, but not earlier (neural crest) or later (mature Schwann cell), results in colony-forming cells capable of multilineage differentiation. In vivo, inactivation of Nf1 using a DhhCre driver beginning at E12.5 elicits plexiform neurofibromas, dermal neurofibromas, and pigmentation. Tumor Schwann cells uniquely show biallelic Nf1 inactivation. Peripheral nerve and tumors contain transiently proliferating Schwann cells that lose axonal contact, providing insight into early neurofibroma formation. We suggest that timing of Nf1 mutation is critical for neurofibroma formation.
Collapse
Affiliation(s)
- Jianqiang Wu
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| | - Jon P. Williams
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| | - Tilat A. Rizvi
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| | - Jennifer J. Kordich
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| | - David Witte
- Division of Pathology, Department of Pediatrics, Cincinnati Children’s Hospital, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| | - Dies Meijer
- Departments of Cell Biology and Genetics, Erasmus University Medical Center, 3000DR Rotterdam, Netherlands
| | - Anat O. Stemmer-Rachamimov
- Departments of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Jose A. Cancelas
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
- Hoxworth Blood Center, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| | - Nancy Ratner
- Division of Experimental Hematology and Cancer Biology, University of Cincinnati College of Medicine, Cincinnati, Ohio, 45229
| |
Collapse
|
30
|
Perrin GQ, Li H, Fishbein L, Thomson SA, Hwang MS, Scarborough MT, Yachnis AT, Wallace MR, Mareci TH, Muir D. An orthotopic xenograft model of intraneural NF1 MPNST suggests a potential association between steroid hormones and tumor cell proliferation. J Transl Med 2007; 87:1092-102. [PMID: 17876295 DOI: 10.1038/labinvest.3700675] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are the most aggressive cancers associated with neurofibromatosis type 1 (NF1). Here we report a practical and reproducible model of intraneural NF1 MPNST, by orthotopic xenograft of an immortal human NF1 tumor-derived Schwann cell line into the sciatic nerves of female scid mice. Intraneural injection of the cell line sNF96.2 consistently produced MPNST-like tumors that were highly cellular and showed extensive intraneural growth. These xenografts had a high proliferative index, were angiogenic, had significant mast cell infiltration and rapidly dominated the host nerve. The histopathology of engrafted intraneural tumors was consistent with that of human NF1 MPNST. Xenograft tumors were readily examined by magnetic resonance imaging, which also was used to assess tumor vascularity. In addition, the intraneural proliferation of sNF96.2 cell tumors was decreased in ovariectomized mice, while replacement of estrogen or progesterone restored tumor cell proliferation. This suggests a potential role for steroid hormones in supporting tumor cell growth of this MPNST cell line in vivo. The controlled orthotopic implantation of sNF96.2 cells provides for the precise initiation of intraneural MPNST-like tumors in a model system suitable for therapeutic interventions, including inhibitors of angiogenesis and further study of steroid hormone effects on tumor cell growth.
Collapse
Affiliation(s)
- George Q Perrin
- Department of Neuroscience, College of Medicine, University of Florida, Gainesville, FL 32610-0244, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Lee FY, Wen MC, Wang J. Epithelioid angiosarcoma arising in a deep-seated plexiform schwannoma: a case report and literature review. Hum Pathol 2007; 38:1096-1101. [PMID: 17574948 DOI: 10.1016/j.humpath.2007.02.019] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2006] [Revised: 02/22/2007] [Accepted: 02/27/2007] [Indexed: 11/25/2022]
Abstract
Angiosarcoma developing within a schwannoma is rare, and only 8 cases have been reported in the literature. Its association with a plexiform schwannoma has never been reported. We describe an epithelioid angiosarcoma arising in a plexiform schwannoma of the sciatic nerve in a 73-year-old man not known to have von Recklinghausen disease. Grossly, the tumor forms a long tubular mass with a multinodular growth pattern on the cut surface. Microscopically, these nodules display characteristics of a schwannoma, including Antoni A, Antoni B, and Verocay bodies. Adjacent to these nodules are high-grade anaplastic epithelioid cells growing in solid sheets, cords, and nests with focal vasoformative channels. The anaplastic cells are immunoreactive to CD31, CD34, and factor VIII related antigen, supporting their endothelial differentiation. Local recurrence and multiple pulmonary metastasis occurred 3 months after complete excision of the tumor. This case demonstrates the first occurrence of an epithelioid angiosarcoma in a deep-seated plexiform schwannoma with an aggressive clinical behavior.
Collapse
Affiliation(s)
- Fang-Yi Lee
- Department of Pathology, Taichung Veterans General Hospital, Taichung 40705, Taiwan
| | - Mei-Chin Wen
- Department of Pathology, Taichung Veterans General Hospital, Taichung 40705, Taiwan; College of Biomedicine and Nursing, HungKuang University, Taichung 40705, Taiwan.
| | - John Wang
- Department of Pathology, Taichung Veterans General Hospital, Taichung 40705, Taiwan; College of Biomedicine and Nursing, HungKuang University, Taichung 40705, Taiwan
| |
Collapse
|
32
|
Mahller YY, Vaikunth SS, Currier MA, Miller SJ, Ripberger MC, Hsu YH, Mehrian-Shai R, Collins MH, Crombleholme TM, Ratner N, Cripe TP. Oncolytic HSV and erlotinib inhibit tumor growth and angiogenesis in a novel malignant peripheral nerve sheath tumor xenograft model. Mol Ther 2007; 15:279-86. [PMID: 17235305 DOI: 10.1038/sj.mt.6300038] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Malignant peripheral nerve sheath tumors (MPNSTs), driven in part by hyperactive Ras and epidermal growth factor receptor (EGFR) signaling, are often incurable. Testing of therapeutics for MPNST has been hampered by lack of adequate xenograft models. We previously documented that human MPNST cells are permissive for lytic infection by oncolytic herpes simplex viruses (oHSV). Herein we developed and characterized a xenograft model of human MPNST and evaluated the antitumor effects of oHSV mutants (G207 and hrR3) and the EGFR inhibitor, erlotinib. Additive cytotoxicity of these agents was found in human MPNST cell lines, suggesting that EGFR signaling is not critical for virus replication. Mice bearing human MPNST tumors treated with G207 or hrR3 by intraperitoneal or intratumoral injection showed tumor-selective virus biodistribution, virus replication, and reduced tumor burden. oHSV injection demonstrated more dramatic antitumor activity than erlotinib. Combination therapies showed a trend toward an increased antiproliferative effect. Both oHSV and erlotinib were antiangiogenic as measured by proangiogenic gene expression, effect on endothelial cells and xenograft vessel density. Overall, oHSVs showed highly potent antitumor effects against MPNST xenografts, an effect not diminished by EGFR inhibition. Our data suggest that inclusion of MPNSTs in clinical trials of oHSV is warranted.
Collapse
Affiliation(s)
- Yonatan Y Mahller
- Division of Hematology/Oncology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
✓The authors present a brief and selective history of surgery for peripheral nerve tumors to illustrate how the current understanding of the nature of disease influences the choice of surgical intervention. There was very little understanding of the anatomy and function of peripheral nerves in ancient times; consequently, surgical treatments for peripheral nerve tumors were based on the writings of authorities. The confusion between traumatic neuromas and genuine nerve sheath tumors coupled with the belief that manipulation of a peripheral nerve might be lethal to the patient stifled the development of surgical techniques for the management of nerve tumors in the 18th and 19th centuries. It was not until the 20th century, with an increased understanding of the microscopic anatomy of nerve sheath tumors, that efficacious surgical treatments for these diseases were developed. Continued advances in the understanding of the biology of these tumors will continue to impact their surgical management.
Collapse
Affiliation(s)
- Ciaran J Powers
- Division of Neurosurgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina 27710, USA
| | | |
Collapse
|
34
|
Thomas SL, De Vries GH. Angiogenic Expression Profile of Normal and Neurofibromin-Deficient Human Schwann Cells. Neurochem Res 2007; 32:1129-41. [PMID: 17404841 DOI: 10.1007/s11064-007-9279-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2006] [Accepted: 12/28/2006] [Indexed: 11/30/2022]
Abstract
Peripheral nerve sheath tumors from individuals with Neurofibromatosis Type 1 (NF1) are highly vascular and contain Schwann cells which are deficient in neurofibromin. This study examines the angiogenic expression profile of neurofibromin-deficient human Schwann cells relative to normal human Schwann cells, characterizing both pro-angiogenic and anti-angiogenic factors. Conditioned media from neurofibromin-deficient Schwann cell lines was pro-angiogenic as evidenced by its ability to stimulate endothelial cell proliferation and migration. Using gene array and protein array analysis, we found increased expression of pro-angiogenic factors and decreased expression of anti-angiogenic factors in neurofibromin-deficient Schwann cells relative to normal human Schwann cells. Neurofibromin-deficient Schwann cells also showed increased expression of several growth factor receptors and decreased expression of an integrin. We conclude that neurofibromin-deficient Schwann cells have dysregulated expression of pro-angiogenic factors, anti-angiogenic factors, growth factor receptors, and an integrin. These dysregulated molecules may contribute to the growth and progression of NF1 peripheral nerve sheath tumors.
Collapse
Affiliation(s)
- Stacey L Thomas
- Research Service, Edward Hines Jr. V.A. Hospital, 5th Avenue and Roosevelt Road, Hines, IL 60141, USA
| | | |
Collapse
|
35
|
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
|
36
|
Thomas SL, Deadwyler GD, Tang J, Stubbs EB, Muir D, Hiatt KK, Clapp DW, De Vries GH. Reconstitution of the NF1 GAP-related domain in NF1-deficient human Schwann cells. Biochem Biophys Res Commun 2006; 348:971-80. [PMID: 16908010 DOI: 10.1016/j.bbrc.2006.07.159] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2006] [Accepted: 07/23/2006] [Indexed: 01/07/2023]
Abstract
Schwann cells derived from peripheral nerve sheath tumors from individuals with Neurofibromatosis Type 1 (NF1) are deficient for the protein neurofibromin, which contains a GAP-related domain (NF1-GRD). Neurofibromin-deficient Schwann cells have increased Ras activation, increased proliferation in response to certain growth stimuli, increased angiogenic potential, and altered cell morphology. This study examined whether expression of functional NF1-GRD can reverse the transformed phenotype of neurofibromin-deficient Schwann cells from both benign and malignant peripheral nerve sheath tumors. We reconstituted the NF1-GRD using retroviral transduction and examined the effects on cell morphology, growth potential, and angiogenic potential. NF1-GRD reconstitution resulted in morphologic changes, a 16-33% reduction in Ras activation, and a 53% decrease in proliferation in neurofibromin-deficient Schwann cells. However, NF1-GRD reconstitution was not sufficient to decrease the in vitro angiogenic potential of the cells. This study demonstrates that reconstitution of the NF1-GRD can at least partially reverse the transformation of human NF1 tumor-derived Schwann cells.
Collapse
Affiliation(s)
- Stacey L Thomas
- Research Service, Edward Hines Jr. V.A. Hospital, 5th Avenue and Roosevelt Road, Hines, IL 60141, USA
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Mahller YY, Rangwala F, Ratner N, Cripe TP. Malignant peripheral nerve sheath tumors with high and low Ras-GTP are permissive for oncolytic herpes simplex virus mutants. Pediatr Blood Cancer 2006; 46:745-54. [PMID: 16124003 DOI: 10.1002/pbc.20565] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Malignant peripheral nerve sheath tumors (MPNSTs) occur most frequently in patients with neurofibromatosis type 1 and are often fatal. Current therapy relies upon radical surgical resection, which often fails to completely remove the tumor. To address the need for novel treatment approaches for this disease, we sought to determine if human MPNST-derived cell lines are sensitive to oncolytic Herpes simplex virus (oHSV) infection. Activation of the Ras pathway and its inhibitory effects on protein kinase R (PKR) activation have been shown to dictate cellular permissivity to oHSV mutants. Because NF-1-associated MPNSTs possess inherent hyperactive Ras, we hypothesized these tumors would be ideal therapeutic targets for oHSVs. PROCEDURE Human MPNST-derived cell lines were examined for sensitivity to oHSV-mediated gene transduction, virus replication, cytotoxicity, and apoptosis. These parameters were correlated with PKR activation following oHSV infection and compared with normal human Schwann cells (NHSCs) without hyperactive Ras. RESULTS MPNST-derived cell lines were efficiently transduced, supported virus replication and were killed by the oncolytic HSV mutants, including sporadic MPNSTs without hyperactive Ras. In contrast to the highly sensitive MPNST cell lines, NHSCs did not support mutant virus replication. CONCLUSIONS MPNSTs are susceptible to lysis by oncolytic HSV mutants, regardless of Ras status. Tumor-selective virus replication in MPNST cells appears to be mediated by both cellular expression of ribonucleotide reductase and prevention of eIF2alpha phosphorylation. Virus-induced cytotoxicity of MPNST cell lines was caused by both direct lysis and apoptosis. Our data suggest the use of oncolytic HSV mutants may represent a novel treatment approach for patients with MPNSTs.
Collapse
Affiliation(s)
- Yonatan Y Mahller
- Division of Hematology/Oncology, University of Cincinnati College of Medicine, Cincinnati, Ohio 45229, USA
| | | | | | | |
Collapse
|
38
|
Gottfried ON, Viskochil DH, Fults DW, Couldwell WT. Molecular, genetic, and cellular pathogenesis of neurofibromas and surgical implications. Neurosurgery 2006; 58:1-16; discussion 1-16. [PMID: 16385324 DOI: 10.1227/01.neu.0000190651.45384.8b] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Neurofibromatosis 1 (NF1) is a common autosomal dominant disease characterized by complex and multicellular neurofibroma tumors. Significant advances have been made in the research of the cellular, genetic, and molecular biology of NF1. The NF1 gene was identified by positional cloning. The functions of its protein product, neurofibromin, in RAS signaling and in other signal transduction pathways are being elucidated, and the important roles of loss of heterozygosity and haploinsufficiency in tumorigenesis are better understood. The Schwann cell was discovered to be the cell of origin for neurofibromas, but understanding of a more complicated interplay of multiple cell types in tumorigenesis, specifically recruited heterogeneous cell types such as mast cells and fibroblasts, has important implications for surgical therapy of these tumors. This review summarizes the most recent NF1 and neurofibroma literature describing the pathogenesis and treatment of nerve sheath tumors. Understanding the biological underpinnings of tumorigenesis in NF1 has implications for future surgical and medical management of neurofibromas.
Collapse
Affiliation(s)
- Oren N Gottfried
- Department of Neurosurgery, University of Utah, Salt Lake City, Utah, USA
| | | | | | | |
Collapse
|
39
|
Abstract
Mutations in the NF1 tumor-suppressor gene underlie neurofibromatosis type 1 (NF1), in which patients are predisposed to certain tumors such as neurofibromas and may associate with vascular disorder. Plexiform neurofibromas are slow growing benign tumors that are highly vascular and can progress to malignancy. The development of neurofibromas requires loss of both Nf1 alleles in Schwann cells destined to become neoplastic and may be exacerbated by Nf1 heterozygosity in other non-neoplastic cells. This study tested the hypothesis that Nf1 heterozygosity exaggerates angiogenesis. We found that Nf1 heterozygous mice showed increased neovascularization in both the retina and cornea in response to hypoxia and bFGF, respectively, compared to their wild-type littermates. The increase in corneal neovascularization was associated with heightened endothelial cell proliferation and migration, and increased infiltration of inflammatory cells. In addition, Nf1 heterozygous endothelial cell cultures showed an exaggerated proliferative response to angiogenic factors, particularly to bFGF. These findings support the conclusion that Nf1 heterozygosity in endothelial cells and perhaps inflammatory cells augments angiogenesis, which may promote neurofibroma formation in NF1.
Collapse
Affiliation(s)
- M Wu
- Department of Pediatrics, Division of Neurology, University of Florida, Gainesville, FL 32610, USA.
| | | | | |
Collapse
|
40
|
ROSENBAUM THORSTEN, KIM HAESUNA, BOISSY YINGL, LING BO, RATNER NANCY. Neurofibromin, the Neurofibromatosis Type 1 Ras-GAP, Is Required for Appropriate P0Expression and Myelination. Ann N Y Acad Sci 2006; 883:203-214. [DOI: 10.1111/j.1749-6632.1999.tb08583.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
41
|
Corfas G, Velardez MO, Ko CP, Ratner N, Peles E. Mechanisms and roles of axon-Schwann cell interactions. J Neurosci 2005; 24:9250-60. [PMID: 15496660 PMCID: PMC6730082 DOI: 10.1523/jneurosci.3649-04.2004] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Gabriel Corfas
- Division of Neuroscience, Children's Hospital, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
42
|
Ling BC, Wu J, Miller SJ, Monk KR, Shamekh R, Rizvi TA, DeCourten-Myers G, Vogel KS, DeClue JE, Ratner N. Role for the epidermal growth factor receptor in neurofibromatosis-related peripheral nerve tumorigenesis. Cancer Cell 2005; 7:65-75. [PMID: 15652750 PMCID: PMC2854500 DOI: 10.1016/j.ccr.2004.10.016] [Citation(s) in RCA: 99] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2003] [Revised: 10/08/2004] [Accepted: 10/25/2004] [Indexed: 01/22/2023]
Abstract
Benign neurofibromas and malignant peripheral nerve sheath tumors are serious complications of neurofibromatosis type 1. The epidermal growth factor receptor is not expressed by normal Schwann cells, yet is overexpressed in subpopulations of Nf1 mutant Schwann cells. We evaluated the role of EGFR in Schwann cell tumorigenesis. Expression of EGFR in transgenic mouse Schwann cells elicited features of neurofibromas: Schwann cell hyperplasia, excess collagen, mast cell accumulation, and progressive dissociation of non-myelin-forming Schwann cells from axons. Mating EGFR transgenic mice to Nf1 hemizygotes did not enhance this phenotype. Genetic reduction of EGFR in Nf1(+/-);p53(+/-) mice that develop sarcomas significantly improved survival. Thus, gain- and loss-of-function experiments support the relevance of EGFR to peripheral nerve tumor formation.
Collapse
Affiliation(s)
- Benjamin C. Ling
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
- Department of Neurosurgery, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Jianqiang Wu
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Shyra J. Miller
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Kelly R. Monk
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Rania Shamekh
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | - Tilat A. Rizvi
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
| | | | - Kristine S. Vogel
- The Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, Texas 78229
| | - Jeffrey E. DeClue
- Laboratory of Cellular Oncology, National Cancer Institute, Bethesda, Maryland 20892
| | - Nancy Ratner
- Departments of Cell Biology, Neurobiology, and Anatomy, University of Cincinnati College of Medicine, Cincinnati, Ohio 45267
- Correspondence:
| |
Collapse
|
43
|
Carroll SL, Stonecypher MS. Tumor suppressor mutations and growth factor signaling in the pathogenesis of NF1-associated peripheral nerve sheath tumors. I. The role of tumor suppressor mutations. J Neuropathol Exp Neurol 2004; 63:1115-23. [PMID: 15581179 DOI: 10.1093/jnen/63.11.1115] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Patients with neurofibromatosis type 1 (NF1), a common autosomal dominant tumor predisposition syndrome, develop benign cutaneous, intraneural, and plexiform neurofibromas and malignant peripheral nerve sheath tumors (MPNSTs), an aggressive form of Schwann cell neoplasm that frequently arises from plexiform neurofibromas. Impressive advances have been made in defining the molecular mechanisms responsible for neurofibroma and MPNST tumorigenesis, including the identification of key tumor suppressor gene mutations, an improved understanding of the functions of these tumor suppressors, and the production of transgenic mouse models in which tumor suppressor gene mutations predispose animals to the development of neurofibromas and MPNSTs. It has also become apparent that dysregulated growth factor signaling cooperates with tumor suppressor mutations to promote neurofibroma and MPNST tumorigenesis. In Part I of this two-part review, we consider findings demonstrating that Schwann cells are the primary neoplastic cell type in neurofibromas and MPNSTs and that specific tumor suppressor gene mutations promote the development of these tumors. In Part II, which will be published in a later issue, we will review evidence indicating that inappropriate growth factor signaling contributes to this process by stimulating the proliferation, survival, and migration of Schwann cells whose regulatory mechanisms have been crippled by a loss of tumor suppressor function.
Collapse
Affiliation(s)
- Steven L Carroll
- Division of Neuropathology, Department of Pathology, The University of Alabama School of Medicine, Birmingham, Alabama 35294-0017, USA.
| | | |
Collapse
|
44
|
Lertsburapa T, De Vries GH. In vitro studies of pigment epithelium-derived factor in human Schwann cells after treatment with axolemma-enriched fraction. J Neurosci Res 2004; 75:624-31. [PMID: 14991838 DOI: 10.1002/jnr.20002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Pigment epithelium-derived factor (PEDF) is a multifunctional protein with known anti-angiogenic and trophic properties, capable of promoting the survival and growth of Schwann cells (SC). Normal rat SCs and ganglioneuroma-derived human SCs secrete PEDF. The ability of normal SC to secrete a number of trophic factors is controlled by axonal contact. Normal human Schwann cells (HSC) and malignant peripheral nerve sheath tumors (MPNST) cell lines synthesize and secrete PEDF as determined by reverse transcription PCR analysis for PEDF mRNA, immunocytochemistry, and Western blot analysis for PEDF protein. Two MPNST cell lines secreted higher levels of PEDF than did HSC. A 90.3% decrease in PEDF mRNA and a 29.3% decrease in secreted PEDF were observed after treatment of HSC with axolemma-enriched fraction (AEF, 100 microg/ml), a neuronal membrane fraction of the axonal plasma membrane used with cultured SC to mimic axonal contact in vitro. PEDF levels remained unchanged, however, in MPNST-derived SC conditioned media under the same treatment paradigm. These results suggest that MPNST SC lose the ability to downregulate PEDF upon axonal contact, which is characteristic of HSC. The elevated PEDF levels expressed by MPNST cell lines may serve to promote their proliferation and survival.
Collapse
Affiliation(s)
- Terakeith Lertsburapa
- Department of Cell Biology, Neurobiology and Anatomy, Loyola University of Chicago, Maywood, Illinois, USA
| | | |
Collapse
|
45
|
De Luca A, Bernardini L, Ceccarini C, Sinibaldi L, Novelli A, Giustini S, Daniele I, Calvieri S, Mingarelli R. Fluorescence in situ hybridization analysis of allelic losses involving the long arm of chromosome 17 in NF1-associated neurofibromas. ACTA ACUST UNITED AC 2004; 150:168-72. [PMID: 15066327 DOI: 10.1016/j.cancergencyto.2003.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2003] [Revised: 08/29/2003] [Accepted: 09/02/2003] [Indexed: 12/01/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common autosomal dominant condition associated with germline mutations of the NF1 gene located at chromosome band 17q11.2. Molecular analysis of a number of NF1-specific tumors has shown the inactivation of both NF1 alleles during tumorigenesis, supporting the tumor suppressor hypothesis for the NF1 gene. Using interphase dual-color fluorescence in situ hybridization (FISH) technique on paraffin-embedded tissues, we studied 11 plexiform, 4 cutaneous, and 6 subcutaneous neurofibromas. Cytogenetic analysis was conducted using two probes, one specific for the NF1 region (RP11-229K15) and one for the centromeric region of chromosome 17 as control. No large somatic deletions were found. Only in one of the plexiform neurofibromas loss of a whole chromosome 17 was observed. If we assume that dual-color FISH analysis is sensitive enough to detect the majority of large somatic deletions present, then other mutational mechanisms affecting the NF1 gene are probably involved in neurofibroma formation, and other tumor suppressor genes may play an important role in NF1 tumorigenesis.
Collapse
Affiliation(s)
- Alessandro De Luca
- IRCCS-CSS, San Giovanni Rotondo and CSS-Mendel Institute, Viale Regina Margherita 261, 00198 Rome, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Huang Y, Rangwala F, Fulkerson PC, Ling B, Reed E, Cox AD, Kamholz J, Ratner N. Role of TC21/R-Ras2 in enhanced migration of neurofibromin-deficient Schwann cells. Oncogene 2004; 23:368-78. [PMID: 14724565 PMCID: PMC2854497 DOI: 10.1038/sj.onc.1207075] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The neurofibromatosis type 1 tumor suppressor protein neurofibromin, is a GTPase activating protein for H-, N-, K-, R-Ras and TC21/R-Ras2 proteins. We demonstrate that Schwann cells derived from Nf1-null mice have enhanced chemokinetic and chemotactic migration in comparison to wild-type controls. Surprisingly, this migratory phenotype is not inhibited by a farnesyltransferase inhibitor or dominant-negative (dn) (N17)H-Ras (which inhibits H-, N-, and K-Ras activation). We postulated that increased activity of R-Ras and/or TC21/R-Ras2, due to loss of Nf1, contributes to increased migration. Mouse Schwann cells (MSCs) express R-Ras and TC21/R-Ras2 and their specific guanine exchange factors, C3G and AND-34. Infection of Nf1-null MSCs with a dn(43N)R-Ras adenovirus (to inhibit both R-Ras and TC21/R-Ras2 activation) decreases migration by approximately 50%. Conversely, expression of activated (72L)TC21/R-Ras2, but not activated (38V)R-Ras, increases migration, suggesting a role of TC21/R-Ras2 activation in the migration of neurofibromin-deficient Schwann cells. TC21/R-Ras2 preferentially couples to the phosphatidylinositol 3-kinase (PI3-kinase) and MAP kinase pathways. Treatment with a PI3-kinase or MAP kinase inhibitor reduces Nf1-null Schwann cell migration, implicating these TC21 effectors in Schwann cell migration. These data reveal a key role for neurofibromin regulation of TC21/R-Ras2 in Schwann cells, a cell type critical to NF1 tumor pathogenesis.
Collapse
Affiliation(s)
- Yuan Huang
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, College of Medicine, 3125 Eden Ave., Cincinnati, OH 45267-0521, USA
| | - Fatima Rangwala
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, College of Medicine, 3125 Eden Ave., Cincinnati, OH 45267-0521, USA
| | - Patricia C Fulkerson
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, College of Medicine, 3125 Eden Ave., Cincinnati, OH 45267-0521, USA
| | - Bo Ling
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, College of Medicine, 3125 Eden Ave., Cincinnati, OH 45267-0521, USA
| | - Erin Reed
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, College of Medicine, 3125 Eden Ave., Cincinnati, OH 45267-0521, USA
| | - Adrienne D Cox
- Departments of Radiation Oncology and Pharmacology, CB7512, Lineberger Cancer Center, UNC-CH, Chapel Hill, NC 27599, USA
| | - John Kamholz
- Department of Neurology, Wayne State University, Elliman Building 3206, 421 East Canfield, Detroit, MI 48201, USA
| | - Nancy Ratner
- Department of Cell Biology, Neurobiology and Anatomy, University of Cincinnati, College of Medicine, 3125 Eden Ave., Cincinnati, OH 45267-0521, USA
- Correspondence: N Ratner;
| |
Collapse
|
47
|
Deliveliotis C, Albanis S, Skolarikos A, Varkarakis J, Protogerou V, Tamvakis N, Alargof E. Solitary neurofibroma of the spermatic cord. Int Urol Nephrol 2004; 34:373-5. [PMID: 12899231 DOI: 10.1023/a:1024415016017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We report a case of solitary neurofibroma of the spermatic cord. Such benign tumors arise from perineural and Schwann cells and may be located in various parts of the body, but are rarely observed in the spermatic cord. No clinical or laboratorial manifestations of von Recklinghausen disease (Neurofibromatosis) were identified. Distinct criteria have been established for a diagnosis of von Recklinghausen disease, so that a solitary neurofibroma may not represent this complex. Only a little number of solitary neurofibromas of the spermatic cord are reported in the literature.
Collapse
Affiliation(s)
- Charalampos Deliveliotis
- Department of Urology, Sismanoglio Hospital, Medical School of Kapodistrian, University of Athens, Greece
| | | | | | | | | | | | | |
Collapse
|
48
|
Babovic-Vuksanovic D, Petrovic L, Knudsen BE, Plummer TB, Parisi JE, Babovic S, Platt JL. Survival of Human Neurofibroma in Immunodeficient Mice and Initial Results of Therapy With Pirfenidone. J Biomed Biotechnol 2004; 2004:79-85. [PMID: 15240917 PMCID: PMC548804 DOI: 10.1155/s1110724304308107] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Neurofibromatosis type I is a common tumor predisposing disease in humans. Surgical therapy can be applied only in selected patients with resectable masses. Hence, development of new therapies for this disease is urgent. We used human neurofibroma implants in mice with severe combined immunodeficiency (SCID) as a model to test the toxicity and potential efficacy of pirfenidone, a new therapeutic agent. Two hundred twelve human neurofibromas were transplanted into various locations in 59 experimental animals, and 30 mice with implants received oral pirfenidone for up to six weeks. Survival of neurofibromas in animals treated with pirfenidone was lower than in the control group $(P=.02)$. Tumors did not change histologic appearance or vascularization in response to pirfenidone. Treatment with pirfenidone, a new antifibrotic agent, inhibits survival of some tumors without causing toxicity in animals.
Collapse
Affiliation(s)
- Dusica Babovic-Vuksanovic
- Department of Medical Genetics, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA
- *Dusica Babovic-Vuksanovic:
| | - Lydia Petrovic
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW,
Rochester, MN 55902, USA
| | - Bruce E. Knudsen
- Transplantation Biology Program, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA
| | - Timothy B. Plummer
- Transplantation Biology Program, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA
| | - Joseph E. Parisi
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 First Street SW,
Rochester, MN 55902, USA
| | - Srdan Babovic
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA
| | - Jeffrey L. Platt
- Department of Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA
| |
Collapse
|
49
|
Abstract
Neurofibromatosis 1, an inherited disorder that affects 1/3500 individuals worldwide, predisposes to the development of benign and malignant peripheral nerve sheath tumors. The disorder results from inactivation of one of the NFI genes. The second NFI gene is typically inactivated in Schwann cells during tumor formation. This article reviews the different types of genetic alterations in NFI in both constitutional and tumor tissues and genetic alterations of other genes that may affect tumorigenesis. These studies have provided insight into the genetic basis of both the variable expression of the disorder and of benign and malignant peripheral nerve sheath tumorigenesis.
Collapse
Affiliation(s)
- Karen Stephens
- Departments of Medicine and Laboratory Medicine, University of Washington, 1959 NE Pacific St., Rm I-204, Box 357720, Seattle, WA 98195-7720, USA.
| |
Collapse
|
50
|
Abstract
We studied the expression of angiogenic factors (vascular endothelial growth factor, basic fibroblast growth factor, platelet-derived growth factor and hepatocyte growth factor) in cutaneous neurofibroma samples from patients with neurofibromatosis-1. Immunohistochemical staining and the reverse transcribed polymerase chain reaction (RT-PCR) method demonstrated that vascular endothelial and basic fibroblast growths factor are highly expressed in neurofibroma cells at both the protein and mRNA level. These data suggest that vascular endothelial and basic fibroblast growth factors may contribute to both the angiogenesis and hypervascularity of neurofibromas.
Collapse
Affiliation(s)
- Yasuhiro Kawachi
- Department of Dermatology, Institute of Clinical Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.
| | | | | | | | | |
Collapse
|