1
|
Paria N, Oxendine I, Podeszwa D, Wassell M, Cornelia R, Wise CA, Rios JJ. Molecular Evidence Supporting MEK Inhibitor Therapy in NF1 Pseudarthrosis. J Bone Joint Surg Am 2025; 107:1098-1106. [PMID: 40168468 PMCID: PMC12080365 DOI: 10.2106/jbjs.24.01007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is a genetic condition predisposing children to fracture pseudarthroses. MEK inhibitors are U.S. Food and Drug Administration-approved or are under study for the treatment of malignant pathologies associated with NF1. However, their potential to treat pseudarthrosis is largely unknown. METHODS Primary cells cultured from control bone or fracture pseudarthroses from children with NF1 were treated with vehicle or with the MEK inhibitors trametinib or selumetinib. Gene expression was evaluated with use of transcriptome sequencing (RNAseq), and the activation of the downstream signaling pathway was evaluated with use of western blotting. Results were replicated in an independent cohort of patient fracture pseudarthrosis-derived primary cells. RESULTS Pseudarthrosis samples were reproducibly associated with the reduced expression of gene signatures implicated in osteoblast differentiation, skeletal development, and the formation of the extracellular matrix. The expression of these gene signatures was significantly rescued following treatment with MEK inhibitors and concomitant reduced MEK/ERK (MAPK) pathway activation. CONCLUSIONS Our study identified molecular signatures associated with fracture pseudarthrosis that were rescued with MEK inhibitor treatment. CLINICAL RELEVANCE MEK inhibitors may promote the healing of fracture pseudarthroses in children with NF1.
Collapse
Affiliation(s)
- Nandina Paria
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas
| | - Ila Oxendine
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas
| | - David Podeszwa
- Department of Orthopedics, Scottish Rite for Children, Dallas, Texas
- Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Meghan Wassell
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas
| | - Reuel Cornelia
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas
| | - Carol A. Wise
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas
- Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, Texas
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas
| | - Jonathan J. Rios
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas
- Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, Texas
- McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
2
|
da Costa ICP, Barreto BCT, Barreto LSDC, Cunha KS, Vieira ADCD, de Souza MMG. Investigation of nasal cavity alterations in individuals with neurofibromatosis type 1 using CBCT. J Craniomaxillofac Surg 2024; 52:1491-1496. [PMID: 39322465 DOI: 10.1016/j.jcms.2024.09.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 08/20/2024] [Accepted: 09/06/2024] [Indexed: 09/27/2024] Open
Abstract
OBJECTIVES This study aimed to investigate nasal septum deviation (NSD), nasal bone length (NBL), and the morphology of the middle nasal conchae (MNC) and inferior nasal conchae (INC), as well as their correlations. MATERIALS AND METHODS The sample included 56 cone-beam computed tomography scans divided into two groups: a study group (SG; individuals with NF1; n = 28) and a control group (CG; individuals without NF1; n = 28). NSD, NBL, MNC, and INC classifications were assessed. MNC images were classified as normal, bullous, paradoxical, secondary, and accessory. INC images were classified as normal, lamellar, compact, combined, and bullous. Intra- and interobserver reliability were evaluated. RESULTS SG had a mean NSD of 11.6° (±4.5°) compared with 9.6° (±3.2°) for the CG, showing moderate deviations with no significant difference between groups. SG had a mean NBL of 22.4 mm (±3.4 mm) compared with 22.1 mm (±3.2 mm) for the CG, with a statistically significant difference. Both groups exhibited normal, bullosa, and accessory MNC classifications. SG INC were normal, lamellar, and combined, whereas CG INC were normal and lamellar. There was a weak correlation between NSD and NBL across groups. CONCLUSION Individuals with NF1 showed longer NBL. The weak correlation between NSD and NBL suggested multifactorial influences on these variations. These findings advance our understanding of craniofacial development in NF1 and highlight the need for further research into nasal cavity involvement in this complex genetic disorder.
Collapse
Affiliation(s)
- Ingrid Cristina Pinto da Costa
- Departamento de Odontopediatria e Ortodontia, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Bruna Caroline Tomé Barreto
- Departamento de Odontopediatria e Ortodontia, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Luísa Schubach da Costa Barreto
- Departamento de Odontologia Social e Preventiva (PRECOM), Faculdade de Odontologia, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Karin Soares Cunha
- Departamento de Patologia, Faculdade de Medicina, Universidade Federal Fluminense, Rio de Janeiro, Brazil
| | - Andréa de Castro Domingos Vieira
- Departamento de Patologia e Diagnóstico Oral, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Margareth Maria Gomes de Souza
- Departamento de Odontopediatria e Ortodontia, Faculdade de Odontologia, Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| |
Collapse
|
3
|
Botero V, Tomchik SM. Unraveling neuronal and metabolic alterations in neurofibromatosis type 1. J Neurodev Disord 2024; 16:49. [PMID: 39217323 PMCID: PMC11365184 DOI: 10.1186/s11689-024-09565-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Neurofibromatosis type 1 (OMIM 162200) affects ~ 1 in 3,000 individuals worldwide and is one of the most common monogenetic neurogenetic disorders that impacts brain function. The disorder affects various organ systems, including the central nervous system, resulting in a spectrum of clinical manifestations. Significant progress has been made in understanding the disorder's pathophysiology, yet gaps persist in understanding how the complex signaling and systemic interactions affect the disorder. Two features of the disorder are alterations in neuronal function and metabolism, and emerging evidence suggests a potential relationship between them. This review summarizes neurofibromatosis type 1 features and recent research findings on disease mechanisms, with an emphasis on neuronal and metabolic features.
Collapse
Affiliation(s)
- Valentina Botero
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA
- Department of Neuroscience, Scripps Research, Scripps Florida, Jupiter, FL, USA
- Skaggs School of Chemical and Biological Sciences, Scripps Research, La Jolla, CA, USA
| | - Seth M Tomchik
- Department of Neuroscience and Pharmacology, University of Iowa, Iowa City, IA, USA.
- Stead Family Department of Pediatrics, University of Iowa, Iowa City, IA, 52242, USA.
- Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, 52242, USA.
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA, 52242, USA.
- Hawk-IDDRC, University of Iowa, Iowa City, IA, 52242, USA.
- Department of Neuroscience, Scripps Research, Scripps Florida, Jupiter, FL, USA.
| |
Collapse
|
4
|
Rios JJ, Juan C, Shelton JM, Paria N, Oxendine I, Wassell M, Kidane YH, Cornelia R, Jeffery EC, Podeszwa DA, Conway SJ, Wise CA, Tower RJ. Spatial transcriptomics implicates impaired BMP signaling in NF1 fracture pseudarthrosis in murine and patient tissues. JCI Insight 2024; 9:e176802. [PMID: 38990653 PMCID: PMC11343587 DOI: 10.1172/jci.insight.176802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/13/2024] Open
Abstract
The neurofibromatosis type 1 (NF1) RASopathy is associated with persistent fibrotic nonunions (pseudarthrosis) in human and mouse skeletal tissue. Here, we performed spatial transcriptomics to define the molecular signatures occurring during normal endochondral healing following fracture in mice. Within the control fracture callus, we observed spatially restricted activation of morphogenetic pathways, such as TGF-β, WNT, and BMP. To investigate the molecular mechanisms contributing to Nf1-deficient delayed fracture healing, we performed spatial transcriptomic analysis on a Postn-cre;Nf1fl/- (Nf1Postn) fracture callus. Transcriptional analyses, subsequently confirmed through phospho-SMAD1/5/8 immunohistochemistry, demonstrated a lack of BMP pathway induction in Nf1Postn mice. To gain further insight into the human condition, we performed spatial transcriptomic analysis of fracture pseudarthrosis tissue from a patient with NF1. Analyses detected increased MAPK signaling at the fibrocartilaginous-osseus junction. Similar to that in the Nf1Postn fracture, BMP pathway activation was absent within the pseudarthrosis tissue. Our results demonstrate the feasibility of delineating the molecular and tissue-specific heterogeneity inherent in complex regenerative processes, such as fracture healing, and reconstructing phase transitions representing endochondral bone formation in vivo. Furthermore, our results provide in situ molecular evidence of impaired BMP signaling underlying NF1 pseudarthrosis, potentially informing the clinical relevance of off-label BMP2 as a therapeutic intervention.
Collapse
Affiliation(s)
- Jonathan J. Rios
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
- McDermott Center for Human Growth and Development
- Department of Pediatrics
- Department of Orthopaedic Surgery
- Simmons Comprehensive Cancer Center
| | | | | | - Nandina Paria
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
| | - Ila Oxendine
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
| | - Meghan Wassell
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
| | - Yared H. Kidane
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
| | - Reuel Cornelia
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
| | - Elise C. Jeffery
- Children’s Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - David A. Podeszwa
- Department of Orthopaedic Surgery
- Department of Orthopaedics, Scottish Rite for Children, Dallas, Texas, USA
| | - Simon J. Conway
- Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Carol A. Wise
- Center for Translational Research, Scottish Rite for Children, Dallas, Texas, USA
- McDermott Center for Human Growth and Development
- Department of Pediatrics
- Department of Orthopaedic Surgery
| | - Robert J. Tower
- McDermott Center for Human Growth and Development
- Department of Surgery
| |
Collapse
|
5
|
Perrin S, Protic S, Bretegnier V, Laurendeau I, de Lageneste OD, Panara N, Ruckebusch O, Luka M, Masson C, Maillard T, Coulpier F, Pannier S, Wicart P, Hadj-Rabia S, Radomska KJ, Zarhrate M, Ménager M, Vidaud D, Topilko P, Parfait B, Colnot C. MEK-SHP2 inhibition prevents tibial pseudarthrosis caused by NF1 loss in Schwann cells and skeletal stem/progenitor cells. Sci Transl Med 2024; 16:eadj1597. [PMID: 38924432 DOI: 10.1126/scitranslmed.adj1597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 01/15/2024] [Accepted: 05/24/2024] [Indexed: 06/28/2024]
Abstract
Congenital pseudarthrosis of the tibia (CPT) is a severe pathology marked by spontaneous bone fractures that fail to heal, leading to fibrous nonunion. Half of patients with CPT are affected by the multisystemic genetic disorder neurofibromatosis type 1 (NF1) caused by mutations in the NF1 tumor suppressor gene, a negative regulator of RAS-mitogen-activated protein kinase (MAPK) signaling pathway. Here, we analyzed patients with CPT and Prss56-Nf1 knockout mice to elucidate the pathogenic mechanisms of CPT-related fibrous nonunion and explored a pharmacological approach to treat CPT. We identified NF1-deficient Schwann cells and skeletal stem/progenitor cells (SSPCs) in pathological periosteum as affected cell types driving fibrosis. Whereas NF1-deficient SSPCs adopted a fibrotic fate, NF1-deficient Schwann cells produced critical paracrine factors including transforming growth factor-β and induced fibrotic differentiation of wild-type SSPCs. To counteract the elevated RAS-MAPK signaling in both NF1-deficient Schwann cells and SSPCs, we used MAPK kinase (MEK) and Src homology 2 containing protein tyrosine phosphatase 2 (SHP2) inhibitors. Combined MEK-SHP2 inhibition in vivo prevented fibrous nonunion in the Prss56-Nf1 knockout mouse model, providing a promising therapeutic strategy for the treatment of fibrous nonunion in CPT.
Collapse
Affiliation(s)
- Simon Perrin
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Sanela Protic
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | | | - Ingrid Laurendeau
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | | | - Nicolas Panara
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
| | - Odile Ruckebusch
- Université Paris Est Creteil, INSERM, IMRB, Plateforme de Cytométrie en flux, 94000 Creteil, France
| | - Marine Luka
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Cécile Masson
- Bioinformatics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
| | - Théodora Maillard
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Fanny Coulpier
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Stéphanie Pannier
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Philippe Wicart
- Department of Pediatric Orthopedic Surgery and Traumatology, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | - Smail Hadj-Rabia
- Department of Dermatology, Reference Center for Rare Skin Diseases (MAGEC), Imagine Institute, Necker-Enfants Malades Hospital, AP-HP, Paris Cité University, 75015 Paris, France
| | | | - Mohammed Zarhrate
- INSERM US24/CNRS UAR3633, Paris Cité University, 75015 Paris, France
- Genomics Core Facility, Institut Imagine-Structure Fédérative de Recherche Necker, INSERM U1163, 75015 Paris, France
| | - Mickael Ménager
- Paris Cité University, Imagine Institute, Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, INSERM UMR 1163, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, INSERM UMR 1163, 75015 Paris, France
| | - Dominique Vidaud
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Piotr Topilko
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| | - Béatrice Parfait
- INSERM UMR S1016, Institut Cochin, Université Paris Cité, 75014 Paris, France
- Service de Médecine Génomique des Maladies de Système et d'Organe, Hôpital Cochin, DMU BioPhyGen, Assistance Publique-Hôpitaux de Paris, AP-HP, Centre-Université Paris Cité, F-75014 Paris, France
| | - Céline Colnot
- Université Paris Est Creteil, INSERM, IMRB, 94000 Creteil, France
| |
Collapse
|
6
|
Almuqbil M, Alshaikh FY, Altwaijri W, Baarmah D, Hommady RH, Alshaikh MY, Alammari F, Alhussain M, Almotawa R, Alqarny F, Kashgari A, Alkhodair R, Alkhater JN, Alkhater LN, Alharthi SA, Alsadi MA, AlRumayyan A. Epidemiology and Outcomes of Neurofibromatosis Type 1 (NF-1): Multicenter Tertiary Experience. J Multidiscip Healthc 2024; 17:1303-1314. [PMID: 38533410 PMCID: PMC10964784 DOI: 10.2147/jmdh.s454921] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 03/13/2024] [Indexed: 03/28/2024] Open
Abstract
Purpose The aim of this manuscript was to assess the epidemiology and clinical features of Neurofibromatosis type 1 (NF-1) based on the newly published revised NF-1 diagnostic criteria and to evaluate complications of NF-1 including neurodevelopmental disorders. Patients and methods A retrospective cross-sectional observational study was conducted in the Ministry of National Guard Health Affairs (MNGHA) healthcare organization branches including four tertiary hospitals and 51 primary health care centers in different regions in Saudi Arabia. This study included all patients diagnosed with NF1 using the revised NIH diagnostic criteria published in 2021 that were registered at the electronic medical records (EMR) from 2015 to 2021. Results A total of 184 patients fulfilled the diagnostic criteria and were included in this study. The median age at diagnosis was 11 years (IQR: 4.00-20.25). The most encountered diagnostic criteria in this study were Café-au-lait macules (85.3%), and (42.9%) were found to have two or more neurofibromas with plexiform neurofibroma being the most common subtype (23.36%), approximately (36.4%) of the patient with optic pathway glioma. Nearby (26.6%) of the patients displayed different type of tumors. Iris Lisch nodules were presented in 36.4% of patients at a median age of 12 years (IQR: 9.0-21.8). Cardiovascular abnormality was encountered in 9.8% of the patients. Around 27.7% of the patients reported headache and 11.4% of the patient suffered from different type of epilepsy. Besides, 10.5% of the patients had intellectual disability, 33.8% suffered from communication disorders, and 4.9% patients had ADHD. Conclusion The results of this study will enable practitioners to adopt a more holistic approach and prioritize numerous attributes, which they can subsequently incorporate into their therapeutic methodologies. Furthermore, the identification of these attributes will facilitate an expeditious and accurate diagnosis. Hence, the implementation of intervention during its nascent phase may result in a more advantageous consequence.
Collapse
Affiliation(s)
- Mohammed Almuqbil
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard, Riyadh, Saudi Arabia
- Division of Pediatric Neurology, Department of Pediatrics, King Abdullah Specialist Children’s Hospital (KASCH), National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | | | - Waleed Altwaijri
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard, Riyadh, Saudi Arabia
- Division of Pediatric Neurology, Department of Pediatrics, King Abdullah Specialist Children’s Hospital (KASCH), National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Duaa Baarmah
- Department of Pediatrics, King Abdullah Bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Raid Harb Hommady
- King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard, Riyadh, Saudi Arabia
- Division of Pediatric Neurology, Department of Pediatrics, King Abdullah Specialist Children’s Hospital (KASCH), National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Maryam Yaseen Alshaikh
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | - Fares Alammari
- College of Medicine, Almaarefa University, Riyadh, Saudi Arabia
| | | | | | - Faris Alqarny
- College of Dentistry, King Saud Bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia
| | - Amna Kashgari
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- Department of Radiology, King Abdullah Specialist Children’s Hospital (KASCH), National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| | - Rayan Alkhodair
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard, Riyadh, Saudi Arabia
- Division of Dermatology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Jumanah N Alkhater
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
| | | | - Sawsan A Alharthi
- Division of Dermatology, King Abdulaziz Medical City, Riyadh, Saudi Arabia
| | - Mada Abdulkarim Alsadi
- Division of Otolaryngology, Head and Neck Surgery, King Abdulaziz University Hospital, Riyadh, Saudi Arabia
| | - Ahmed AlRumayyan
- College of Medicine, King Saud Bin Abdulaziz University for Health Sciences (KSAU-HS), Riyadh, Saudi Arabia
- King Abdullah International Medical Research Center (KAIMRC), Ministry of National Guard, Riyadh, Saudi Arabia
- Division of Pediatric Neurology, Department of Pediatrics, King Abdullah Specialist Children’s Hospital (KASCH), National Guard Health Affairs (NGHA), Riyadh, Saudi Arabia
| |
Collapse
|
7
|
Kaspiris A, Vasiliadis E, Iliopoulos ID, Panagopoulos F, Melissaridou D, Lianou I, Ntourantonis D, Savvidou OD, Papadimitriou E, Pneumaticos SG. Bone mineral density, vitamin D and osseous metabolism indices in neurofibromatosis type 1: A systematic review and meta-analysis. Bone 2024; 180:116992. [PMID: 38141750 DOI: 10.1016/j.bone.2023.116992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/10/2023] [Accepted: 12/18/2023] [Indexed: 12/25/2023]
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is a genetic autosomal neurocutaneous syndrome correlated with skeletal dysplasia and defects in the osseous microarchitecture. The physiological mechanism for the development of NF1-related bone abnormal turnover is still unclear. OBJECTIVES A meta-analysis was performed to investigate the effects of NF1 on bone mineral density (BMD) and osseous metabolic indices in order to provide clinical evidence for the pathogenesis of the associated skeletal deformities. METHODS A systematic literature review search was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines in the PubMed/Medline and Web of Science databases from the date of inception of each database through to 10 September 2023. Specific inclusion and exclusion criteria were applied for the identification of studies examining the effects of NF1 on bone strength and metabolism. The Newcastle-Ottawa and Jadad scales were applied to assess the quality of the included studies. RevMan 5.3 software was used for the analysis of the data, and MedCalc was applied to examine publication bias. RESULTS Overall, 13 studies met the inclusion criteria comprised of 5 cross-sectional, 6 case-control and 2 retrospective studies. 703 patients and 973 healthy subjects formed the NF1 and control group, respectively. The results of the meta-analysis displayed that lumbar (SMD = -3.85, 95%CI = -7.53 to -0.18, Z = 2.05, p = 0.04) and femoral (SMD = -4.78, 95%CI = -8.86 to -0.69, Z = 2.29, p = 0.02) BMD was reduced in the NF1 group. Both in children and adults the serum levels of 25 hydroxyvitamin D3 were also decreased in NF1 group, but without any statistical significance (SMD = -0.62, 95%CI = -1.34 to -0.11, Z = 1.66, p = 0.10). Serum Parathyroid hormone (PTH) (SMD = 0.73, 95%CI = 0.31 to 1.15, Z = 3.43, p = 0.0006) and C-telopeptide of type 1 collagen (CTX) (SMD = 0.82, 95%CI = 0.33 to 1.30, Z = 3.29, p = 0.001) were elevated in NF1 patients, while serum calcium (SMD = -0.10, 95%CI = -0.74 to 0.53, Z = 0.32, p = 0.75) phosphorous (SMD = 0.33, 95%CI = -0.38 to 1.05, Z = 0.92, p = 0.36), alkaline phosphatase (ALP) (SMD = -0.36, 95%CI = -0.77 to 0.05, Z = 1.71, p = 0.09), osteocalcin (SMD = 1.81, 95%CI = -0.37 to -3.98, Z = 1.63, p = 0.10) and bone formation markers (SMD = 0.28, 95%CI = -0.37 to -0.94, Z = 0.85, p = 0.39) were not. CONCLUSION NF1 is associated with decreased BMD at the lumbar spine and femur. Taking into account that the serum levels of PTH, CTX were increased whereas the concentrations of vitamin D, calcium, phosphorous, ALP, osteocalcin and bone formation markers were not altered significantly in the NF1 patients compared with the healthy subjects, a vitamin D independent dysregulated bone cellular activity could be considered. STUDY REGISTRATION Registered on PROSPERO (CRD42023424751).
Collapse
Affiliation(s)
- Angelos Kaspiris
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Patras 26504, Greece.
| | - Elias Vasiliadis
- Third Department of Orthopaedic Surgery, "KAT" General Hospital and Medical School, University of Athens, Athens 14561, Greece
| | - Ilias D Iliopoulos
- Department of Orthopaedic Surgery, "Rion" University Hospital and Medical School, School of Health Sciences, University of Patras, Patras 26504,Greece
| | - Fotis Panagopoulos
- Department of Orthopaedic Surgery, General Hospital of Eastern Achaia-NHS, Aigion, Greece
| | - Dimitra Melissaridou
- First Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, "ATTIKON" University General Hospital, Rimini 1, Athens 12462, Greece
| | - Ioanna Lianou
- Department of Orthopaedic Surgery, "Rion" University Hospital and Medical School, School of Health Sciences, University of Patras, Patras 26504,Greece
| | - Dimitrios Ntourantonis
- Emergency Department, Rion" University Hospital and Medical School, School of Health Sciences, University of Patras, Patras 26504, Greece
| | - Olga D Savvidou
- First Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, "ATTIKON" University General Hospital, Rimini 1, Athens 12462, Greece
| | - Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Department of Pharmacy, School of Health Sciences, University of Patras, Patras 26504, Greece
| | - Spiros G Pneumaticos
- Third Department of Orthopaedic Surgery, "KAT" General Hospital and Medical School, University of Athens, Athens 14561, Greece
| |
Collapse
|
8
|
Charoenngam N, Wattanachayakul P, Jaroenlapnopparat A, Ungprasert P, Chenbhanich J. Bone Mineral Density in Neurofibromatosis Type 1: A Systematic Review and Meta-Analysis. Calcif Tissue Int 2023:10.1007/s00223-023-01094-z. [PMID: 37221347 DOI: 10.1007/s00223-023-01094-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 04/29/2023] [Indexed: 05/25/2023]
Abstract
To assess BMD in patients with neurofibromatosis type 1 (NF1) using systematic review and meta-analysis technique. Potentially eligible studies were identified from Medline and EMBASE databases from inception to February 2023 using search strategy that comprised terms for "Bone mineral density" and Neurofibromatosis type 1″. Eligible study must include adult or pediatric patients with NF1. The study must report mean Z-score with variance of total body, lumbar spine, femoral neck or total hip BMD of the studied patients. Point estimates with standard errors were retrieved from each study and were combined using the generic inverse variance method. A total of 1,165 articles were identified. After systematic review, 19 studies were included. The meta-analysis revealed that patients with NF1 had negative mean Z-scores for total body BMD (pooled mean Z-score -0.808; 95%CI, -1.025 to -0.591) and BMD at lumbar spine (pooled mean Z-score -1.104; 95%CI, -1.376 to -0.833), femoral neck (pooled mean Z-score -0.726; 95%CI, -0.893 to -0.560) and total hip (pooled mean Z-score -1.126; 95%CI, -2.078 to -0.173). The subgroup meta-analysis in pediatric patients aged < 18 years revealed that patients with NF1 had negative mean Z-scores for lumbar spine BMD (pooled mean Z-score -0.938; 95%CI, -1.299 to -0.577) and femoral neck BMD (pooled mean Z-score -0.585; 95%CI, -0.872 to -0.298). The current meta-analysis found that patients with NF1 had low Z-scores although the degree of low BMD may not be of clinical significance. The results do not support the role of early BMD screening in children and young adults with NF1.
Collapse
Affiliation(s)
- Nipith Charoenngam
- Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand.
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, 330 Mt Auburn St, Cambridge, MA, 02138, USA.
| | | | | | - Patompong Ungprasert
- Department of Rheumatic and Immunologic Diseases, Cleveland Clinic, Cleveland, OH, USA
| | - Jirat Chenbhanich
- Department of Genetics and Genomic Sciences, Case Western Reserve University, Cleveland, OH, USA
| |
Collapse
|
9
|
Chelleri C, Scala M, De Marco P, Guerriero V, Ognibene M, Madia F, Guerrisi S, Di Duca M, Torre M, Tamburro S, Scudieri P, Piccolo G, Mattioli G, Buffelli F, Uva P, Vozzi D, Fulcheri E, Striano P, Diana MC, Zara F. Somatic Double Inactivation of NF1 Associated with NF1-Related Pectus Excavatum Deformity. Hum Mutat 2023; 2023:3160653. [PMID: 40225171 PMCID: PMC11918561 DOI: 10.1155/2023/3160653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/15/2023] [Accepted: 04/04/2023] [Indexed: 04/15/2025]
Abstract
Neurofibromatosis type 1 (NF1) is a neurocutaneous genetic disorder with a broad spectrum of associated signs and symptoms, including skeletal anomalies. The association of NF1 with anterior chest wall deformities has been recently reported, especially the pectus excavatum (PE). Over the years, several authors have suggested loss of heterozygosity (LOH) as the possible pathogenic mechanism underlying the development of the typical NF1 skeletal features. Here, we report a NF1 patient with severe chest deformity and harboring the germline heterozygous pathogenic NF1 variant NM_001042492.3: c.4271delC p.(Ala1424Glufs∗4). Through next-generation sequencing (NGS), we investigated the affected cartilage from the PE deformity and identified the additional frameshift variant NM_001042492.3: c.2953delC p.(Gln985Lysfs∗7), occurring as a somatic NF1 second hit mutation. Exome sequencing confirmed the absence of additional variants of potential pathogenic relevance. Western blot analysis showed the absence of wild-type NF1 protein in the cartilage of the patient, consistent with a somatic double inactivation (SDI) of NF1. Taken together, our findings support the role of SDI in NF1-related PE, widening the spectrum of the pathophysiological mechanisms involved in NF1-related skeletal features.
Collapse
Affiliation(s)
- Cristina Chelleri
- Pediatric Neurology and Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Marcello Scala
- Pediatric Neurology and Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Patrizia De Marco
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Vittorio Guerriero
- Pediatric Thoracic and Airway Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Pediatric Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marzia Ognibene
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Francesca Madia
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Sara Guerrisi
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Marco Di Duca
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Michele Torre
- Pediatric Thoracic and Airway Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Pediatric Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Serena Tamburro
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Paolo Scudieri
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Gianluca Piccolo
- Pediatric Neurology and Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Girolamo Mattioli
- Pediatric Thoracic and Airway Surgery Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | - Paolo Uva
- Genomic Facility, Istituto Italiano di Tecnologia, Genova, Italy
| | - Diego Vozzi
- Unità di Bioinformatica Clinica, Direzione Scientifica, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Ezio Fulcheri
- Clinical Pathology Unit, IRCCS Istituto Giannina Gaslini, Italy
| | - Pasquale Striano
- Pediatric Neurology and Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DINOGMI), University of Genoa, Genoa, Italy
| | - Maria Cristina Diana
- Pediatric Neurology and Neuromuscular Disorders Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Federico Zara
- Medical Genetics Unit, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| |
Collapse
|
10
|
Lu Z, Chen P, Xu Q, Li B, Jiang S, Jiang L, Zheng X. Constitutive and conditional gene knockout mice for the study of intervertebral disc degeneration: Current status, decision considerations, and future possibilities. JOR Spine 2023; 6:e1242. [PMID: 36994464 PMCID: PMC10041386 DOI: 10.1002/jsp2.1242] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 12/13/2022] [Accepted: 12/22/2022] [Indexed: 01/09/2023] Open
Abstract
There have been an increasing number of patients with degenerative disc diseases due to the aging population. In light of this, studies on the pathogenesis of intervertebral disc degeneration have become a hot topic, and gene knockout mice have become a valuable tool in this field of research. With the development of science and technology, constitutive gene knockout mice can be constructed using homologous recombination, zinc finger nuclease, transcription activator-like effector nuclease technology and clustered regularly interspaced short palindromic repeats/Cas9 (CRISPR/Cas9) system, and conditional gene knockout mice can be constructed using the Cre/LoxP system. The gene-edited mice using these techniques have been widely used in the studies on disc degeneration. This paper reviews the development process and principles of these technologies, functions of the edited genes in disc degeneration, advantages, and disadvantages of different methods and possible targets of the specific Cre recombinase in intervertebral discs. Recommendations for the choice of suitable gene-edited model mice are presented. At the same time, possible technological improvements in the future are also discussed.
Collapse
Affiliation(s)
- Ze‐Yu Lu
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Peng‐Bo Chen
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Qing‐Yin Xu
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Bo Li
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Sheng‐Dan Jiang
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Lei‐Sheng Jiang
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xin‐Feng Zheng
- Spine CenterXinhua Hospital, Shanghai Jiao Tong University School of MedicineShanghaiChina
| |
Collapse
|
11
|
Uvaraj NR, Bosco A. Scoliosis in Neurofibromatosis. PAEDIATRIC SCOLIOSIS 2023:557-581. [DOI: 10.1007/978-981-99-3017-3_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
12
|
Stevenson DA, Viscogliosi G, Leoni C. Bone health in RASopathies. AMERICAN JOURNAL OF MEDICAL GENETICS. PART C, SEMINARS IN MEDICAL GENETICS 2022; 190:459-470. [PMID: 36461161 DOI: 10.1002/ajmg.c.32020] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 11/07/2022] [Accepted: 11/18/2022] [Indexed: 12/04/2022]
Abstract
The RASopathies are a group of disorders due to pathogenic variants in genes involved in the Ras/MAPK pathway, many of which have overlapping clinical features (e.g., neurofibromatosis type 1, Costello syndrome, cardiofaciocutaneous syndrome and Noonan syndrome) including musculoskeletal manifestations. Osteopenia and osteoporosis are reported in many of the RASopathies suggesting a shared pathogenesis. Even though osteopenia and osteoporosis are often detected and fractures have been reported, the clinical impact of bone mineralization defects on the skeleton of the various syndromes is poorly understood. Further knowledge of the role of the Ras/MAPK pathway on the bone cellular function, and more detailed musculoskeletal phenotyping will be critical in helping to develop therapies to improve bone health in the RASopathies.
Collapse
Affiliation(s)
- David A Stevenson
- Department of Pediatrics, Division of Medical Genetics, Stanford University, Stanford, California, USA
| | - Germana Viscogliosi
- Center for Rare Diseases and Birth Defect, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Chiara Leoni
- Center for Rare Diseases and Birth Defect, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| |
Collapse
|
13
|
Ahmed R, Uppuganti S, Derasari S, Meyer J, Pennings JS, Elefteriou F, Nyman JS. Identifying Bone Matrix Impairments in a Mouse Model of Neurofibromatosis Type 1 (NF1) by Clinically Translatable Techniques. J Bone Miner Res 2022; 37:1603-1621. [PMID: 35690920 PMCID: PMC9378557 DOI: 10.1002/jbmr.4633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 05/25/2022] [Accepted: 06/04/2022] [Indexed: 11/06/2022]
Abstract
Three-to-four percent of children with neurofibromatosis type 1 (NF1) present with unilateral tibia bowing, fracture, and recalcitrant healing. Alkaline phosphatase (ALP) enzyme therapy prevented poor bone mineralization and poor mechanical properties in mouse models of NF1 skeletal dysplasia; but transition to clinical trials is hampered by the lack of a technique that (i) identifies NF1 patients at risk of tibia bowing and fracture making them eligible for trial enrollment and (ii) monitors treatment effects on matrix characteristics related to bone strength. Therefore, we assessed the ability of matrix-sensitive techniques to provide characteristics that differentiate between cortical bone from mice characterized by postnatal loss of Nf1 in Osx-creTet-Off ;Nf1flox/flox osteoprogenitors (cKO) and from wild-type (WT) mice. Following euthanasia at two time points of bone disease progression, femur and tibia were harvested from both genotypes (n ≥ 8/age/sex/genotype). A reduction in the mid-diaphysis ultimate force during three-point bending at 20 weeks confirmed deleterious changes in bone induced by Nf1 deficiency, regardless of sex. Pooling females and males, low bound water (BW), and low cortical volumetric bone mineral density (Ct.vBMD) were the most accurate outcomes in distinguishing cKO from WT femurs with accuracy improving with age. Ct.vBMD and the average unloading slope (Avg-US) from cyclic reference point indentation tests were the most sensitive in differentiating WT from cKO tibias. Mineral-to-matrix ratio and carbonate substitution from Raman spectroscopy were not good classifiers. However, when combined with Ct.vBMD and BW (femur), they helped predict bending strength. Nf1 deficiency in osteoprogenitors negatively affected bone microstructure and matrix quality with deficits in properties becoming more pronounced with duration of Nf1 deficiency. Clinically measurable without ionizing radiation, BW and Avg-US are sensitive to deleterious changes in bone matrix in a preclinical model of NF1 bone dysplasia and require further clinical investigation as potential indicators of an onset of bone weakness in children with NF1. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Rafay Ahmed
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sasidhar Uppuganti
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shrey Derasari
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Joshua Meyer
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Jacquelyn S Pennings
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Musculoskeletal Research, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Florent Elefteriou
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Department of Orthopaedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Jeffry S Nyman
- Department of Orthopaedic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.,Center for Bone Biology, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA.,Center for Musculoskeletal Research, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| |
Collapse
|
14
|
Georgieva VS, Bluhm B, Probst K, Zhu M, Heilig J, Niehoff A, Brachvogel B. Ablation of the miRNA cluster 24 in cartilage and osteoblasts impairs bone remodeling. Sci Rep 2022; 12:9116. [PMID: 35650319 PMCID: PMC9160244 DOI: 10.1038/s41598-022-13231-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 05/13/2022] [Indexed: 11/28/2022] Open
Abstract
MicroRNAs (miRNAs) post-transcriptionally regulate cartilage and bone development and function, however, only few miRNAs have been described to play a role for cartilage to bone transition in vivo. Previously, we showed that cartilage-specific deletion of the Mirc24 cluster in newborn male mice leads to impaired growth plate cartilage development due to increased RAF/MEK/ERK signaling and affects the stability of the cartilage extracellular matrix on account of decreased SOX6 and SOX9 and increased MMP13 levels. Here, we studied how Mirc24 cluster inactivation in cartilage and osteoblasts leads to an increased bone density associated with defects in collagen remodeling in trabecular bone. No changes in osteoblast distribution were observed, whereas the number of osteoclasts was reduced and TRAP activity in osteoclasts decreased. Surprisingly, an increased level of cluster-encoded miR-322 or miR-503 raises Rankl gene expression and inactivation of the cluster in chondrocytes reduces Rankl expression. These results suggest that the Mirc24 cluster regulates Rankl expression in chondrocytes at the chondro-osseous border, where the cluster is mainly expressed to modulate osteoclast formation, bone remodeling and bone integrity.
Collapse
Affiliation(s)
- Veronika S Georgieva
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Björn Bluhm
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Kristina Probst
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Mengjie Zhu
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany
| | - Juliane Heilig
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, 50933, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, 50931, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics, German Sport University Cologne, 50933, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics (CCMB), University of Cologne, 50931, Cologne, Germany
| | - Bent Brachvogel
- Center for Biochemistry, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany. .,Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, 50931, Cologne, Germany.
| |
Collapse
|
15
|
Neifert SN, Khan HA, Kurland DB, Kim NC, Yohay K, Segal D, Samdani A, Hwang S, Lau D. Management and surgical outcomes of dystrophic scoliosis in neurofibromatosis type 1: a systematic review. Neurosurg Focus 2022; 52:E7. [PMID: 35535821 DOI: 10.3171/2022.2.focus21790] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 02/22/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVE
Neurofibromatosis type 1 (NF1) dystrophic scoliosis is an early-onset, rapidly progressive multiplanar deformity. There are few studies on the surgical management of this patient population. Specifically, perioperative morbidity, instrument-related complications, and quality-of-life outcomes associated with surgical management have not been systematically evaluated. In this study, the authors aimed to perform a systematic review on the natural history, management options, and surgical outcomes in patients who underwent NF1 dystrophic scoliosis surgery.
METHODS
A PubMed search for articles with “neurofibromatosis” and either “dystrophic” or “scoliosis” in the title or abstract was performed. Articles with 10 or more patients undergoing surgery for NF1 dystrophic scoliosis were included. Data regarding indications, treatment details, morbidity, and outcomes were summarized and analyzed with descriptive statistics.
RESULTS
A total of 310 articles were identified, 48 of which were selected for full-text review; 30 studies describing 761 patients met the inclusion criteria. The mean age ranged from 7 to 22 years, and 99.7% of patients were younger than 18 years. The mean preoperative coronal Cobb angle was 75.2°, and the average correction achieved was 40.3°. The mean clinical follow-up in each study was at least 2 years (range 2.2–19 years). All patients underwent surgery with the intent of deformity correction. The scoliosis regions addressed were thoracic curves (69.6%) and thoracolumbar (11.1%) and lumbar (14.3%) regions. The authors reported on a variety of approaches: posterior-only, combined anterior-posterior, and growth-friendly surgery. For fixation techniques, 42.5% of patients were treated with hybrid constructs, 51.5% with pedicle screw–only constructs, and 6.0% with hook-based constructs. Only 0.9% of patients underwent a vertebral column resection. The nonneurological complication rate was 14.0%, primarily dural tears and wound infections. The immediate postoperative neurological deficit rate was 2.1%, and the permanent neurological deficit rate was 1.2%. Ultimately, 21.5% required revision surgery, most commonly for implant-related complications. Loss of correction in both the sagittal and coronal planes commonly occurred at follow-up. Five papers supplied validated patient-reported outcome measures, showing improvement in the mental health, self-image, and activity domains.
CONCLUSIONS
Data on the surgical outcomes of dystrophic scoliosis correction are heterogeneous and sparse. The perioperative complication rate appears to be high, although reported rates of neurological deficits appear to be lower than clinically observed and may be underreported. The incidence of implant-related failures requiring revision surgery is high. There is a great need for multicenter prospective studies of this complex type of deformity.
Collapse
Affiliation(s)
- Sean N. Neifert
- Department of Neurological Surgery, New York University, New York, New York
| | - Hammad A. Khan
- Department of Neurological Surgery, New York University, New York, New York
| | - David B. Kurland
- Department of Neurological Surgery, New York University, New York, New York
| | - Nora C. Kim
- Department of Neurological Surgery, New York University, New York, New York
| | - Kaleb Yohay
- Department of Neurology and Comprehensive Neurofibromatosis Center, New York University, New York, New York; and
| | - Devorah Segal
- Department of Neurology and Comprehensive Neurofibromatosis Center, New York University, New York, New York; and
| | - Amer Samdani
- Shriners Hospital for Children, Philadelphia, Pennsylvania
| | - Steven Hwang
- Shriners Hospital for Children, Philadelphia, Pennsylvania
| | - Darryl Lau
- Department of Neurological Surgery, New York University, New York, New York
| |
Collapse
|
16
|
Greenblatt MB, Shim JH, Bok S, Kim JM. The Extracellular Signal-Regulated Kinase Mitogen-Activated Protein Kinase Pathway in Osteoblasts. J Bone Metab 2022; 29:1-15. [PMID: 35325978 PMCID: PMC8948490 DOI: 10.11005/jbm.2022.29.1.1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 02/17/2022] [Indexed: 12/01/2022] Open
Abstract
Extracellular signal-regulated kinases (ERKs) are evolutionarily ancient signal transducers of the mitogen-activated protein kinase (MAPK) family that have long been linked to the regulation of osteoblast differentiation and bone formation. Here, we review the physiological functions, biochemistry, upstream activators, and downstream substrates of the ERK pathway. ERK is activated in skeletal progenitors and regulates osteoblast differentiation and skeletal mineralization, with ERK serving as a key regulator of Runt-related transcription factor 2, a critical transcription factor for osteoblast differentiation. However, new evidence highlights context-dependent changes in ERK MAPK pathway wiring and function, indicating a broader set of physiological roles associated with changes in ERK pathway components or substrates. Consistent with this importance, several human skeletal dysplasias are associated with dysregulation of the ERK MAPK pathway, including neurofibromatosis type 1 and Noonan syndrome. The continually broadening array of drugs targeting the ERK pathway for the treatment of cancer and other disorders makes it increasingly important to understand how interference with this pathway impacts bone metabolism, highlighting the importance of mouse studies to model the role of the ERK MAPK pathway in bone formation.
Collapse
Affiliation(s)
- Matthew B. Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical, New York, NY,
USA
- Research Division, Hospital for Special Surgery, New York, NY,
USA
| | - Jae-Hyuck Shim
- Division of Rheumatology, Department of Medicine, UMass Chan Medical School, Worcester, MA,
USA
- Horae Gene Therapy Center, and Li Weibo Institute for Rare Diseases Research, UMass Chan Medical School, Worcester, MA,
USA
| | - Seoyeon Bok
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical, New York, NY,
USA
| | - Jung-Min Kim
- Division of Rheumatology, Department of Medicine, UMass Chan Medical School, Worcester, MA,
USA
| |
Collapse
|
17
|
Mo J, Moye SL, McKay RM, Le LQ. Neurofibromin and suppression of tumorigenesis: beyond the GAP. Oncogene 2022; 41:1235-1251. [PMID: 35066574 PMCID: PMC9063229 DOI: 10.1038/s41388-021-02156-y] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/01/2021] [Accepted: 12/13/2021] [Indexed: 12/15/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disease and one of the most common inherited tumor predisposition syndromes, affecting 1 in 3000 individuals worldwide. The NF1 gene encodes neurofibromin, a large protein with RAS GTP-ase activating (RAS-GAP) activity, and loss of NF1 results in increased RAS signaling. Neurofibromin contains many other domains, and there is considerable evidence that these domains play a role in some manifestations of NF1. Investigating the role of these domains as well as the various signaling pathways that neurofibromin regulates and interacts with will provide a better understanding of how neurofibromin acts to suppress tumor development and potentially open new therapeutic avenues. In this review, we discuss what is known about the structure of neurofibromin, its interactions with other proteins and signaling pathways, its role in development and differentiation, and its function as a tumor suppressor. Finally, we discuss the latest research on potential therapeutics for neurofibromin-deficient neoplasms.
Collapse
Affiliation(s)
- Juan Mo
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Stefanie L Moye
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Renee M McKay
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA
| | - Lu Q Le
- Department of Dermatology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- UTSW Comprehensive Neurofibromatosis Clinic, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
- O'Donnell Brain Institute, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, 75390-9069, USA.
| |
Collapse
|
18
|
Leoni C, Bisanti C, Viscogliosi G, Onesimo R, Massese M, Giorgio V, Corbo F, Acampora A, Cipolla C, Flex E, Dell'Atti C, Rigante D, Tartaglia M, Zampino G. Bone tissue homeostasis and risk of fractures in Costello syndrome: A 4-year follow-up study. Am J Med Genet A 2022; 188:422-430. [PMID: 34913244 DOI: 10.1002/ajmg.a.62615] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Revised: 10/04/2021] [Accepted: 11/28/2021] [Indexed: 12/21/2022]
Abstract
Costello syndrome (CS) is a neurodevelopmental disorder with a distinctive musculoskeletal phenotype and reduced bone mineral density (BMD) caused by activating de novo mutations in the HRAS gene. Herein, we report the results of a prospective study evaluating the efficacy of a 4-year vitamin D supplementation on BMD and bone health. A cohort of 16 individuals ranging from pediatric to adult age with molecularly confirmed CS underwent dosages of bone metabolism biomarkers (serum/urine) and dual-energy X-ray absorptiometry (DXA) scans to assess bone and body composition parameters. Results were compared to age-matched control groups. At baseline evaluation, BMD was significantly reduced (p ≤ 0.05) compared to controls, as were the 25(OH)vitD levels. Following the 4-year time interval, despite vitamin D supplementation therapy at adequate dosages, no significant improvement in BMD was observed. The present data confirm that 25(OH)vitD and BMD parameters are reduced in CS, and vitamin D supplementation is not sufficient to restore proper BMD values. Based on this evidence, routine monitoring of bone homeostasis to prevent bone deterioration and possible fractures in adult patients with CS is highly recommended.
Collapse
Affiliation(s)
- Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristian Bisanti
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Germana Viscogliosi
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Miriam Massese
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valentina Giorgio
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Corbo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Acampora
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Clelia Cipolla
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Claudia Dell'Atti
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Donato Rigante
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
19
|
Leoni C, Viscogliosi G, Onesimo R, Bisanti C, Massese M, Giorgio V, Corbo F, Tedesco M, Acampora A, Cipolla C, Dell'Atti C, Flex E, Gervasoni J, Primiano A, Rigante D, Tartaglia M, Zampino G. Characterization of bone homeostasis in individuals affected by cardio-facio-cutaneous syndrome. Am J Med Genet A 2022; 188:414-421. [PMID: 34854525 DOI: 10.1002/ajmg.a.62588] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 11/11/2021] [Accepted: 11/16/2021] [Indexed: 01/10/2023]
Abstract
Cardio-facio-cutaneous syndrome (CFCS) is a rare disorder characterized by distinctive craniofacial appearance, cardiac, neurologic, cutaneous, and musculoskeletal abnormalities. It is due to heterozygous mutations in BRAF, MAP2K1, MAP2K2, and KRAS genes, belonging to the RAS/MAPK pathway. The role of RAS signaling in bone homeostasis is highly recognized, but data on bone mineral density (BMD) in CFCS are lacking. In the present study we evaluated bone parameters, serum and urinary bone metabolites in 14 individuals with a molecularly confirmed diagnosis of CFCS. Bone assessment was performed through dual X-ray absorptiometry (DXA); height-adjusted results were compared to age- and sex-matched controls. Blood and urinary bone metabolites were also analyzed and compared to the reference range. Despite vitamin D supplementation and almost normal bone metabolism biomarkers, CFCS patients showed significantly decreased absolute values of DXA-assessed subtotal and lumbar BMD (p ≤ 0.05), compared to controls. BMD z-scores and t-scores (respectively collected for children and adults) were below the reference range in CFCS, while normal in healthy controls. These findings confirmed a reduction in BMD in CFCS and highlighted the importance of monitoring bone health in these affected individuals.
Collapse
Affiliation(s)
- Chiara Leoni
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Germana Viscogliosi
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Roberta Onesimo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Cristian Bisanti
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Miriam Massese
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Valentina Giorgio
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Corbo
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Marta Tedesco
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Anna Acampora
- Section of Hygiene, University Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Clelia Cipolla
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Claudia Dell'Atti
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario "A. Gemelli" IRCCS, Rome, Italy
| | - Elisabetta Flex
- Department of Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Jacopo Gervasoni
- Department of Laboratory and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Aniello Primiano
- Department of Laboratory and Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Donato Rigante
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore, Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giuseppe Zampino
- Center for Rare Diseases and Birth Defects, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Università Cattolica Sacro Cuore, Rome, Italy
| |
Collapse
|
20
|
Prevalence and characteristics of systemic conditions in patients undergoing orthognathic surgery: a retrospective study. Int J Oral Maxillofac Surg 2022; 51:1205-1210. [PMID: 35221148 DOI: 10.1016/j.ijom.2022.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 11/21/2021] [Accepted: 02/04/2022] [Indexed: 11/20/2022]
|
21
|
Li H, Zhang W, Yao Z, Guo R, Hao C, Zhang X. Genotypes and clinical intervention of patients with neurofibromatosis type 1 associated dystrophic scoliosis. Front Pediatr 2022; 10:918136. [PMID: 36061378 PMCID: PMC9434403 DOI: 10.3389/fped.2022.918136] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 07/27/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE To analyze the genotypic characteristics of patients with neurofibromatosis type 1 (NF1) associated dystrophic scoliosis and to summarize the outcomes of the surgical treatment of these patients. METHODS Exome sequencing (ES) combined with multiplex ligation-dependent probe amplification (MLPA) was used for genotypic identification. All patients underwent surgical treatments for spinal deformities, and the outcomes of the surgery was summarized by analyzing the clinical and imaging parameters before and after the surgery. RESULTS Fourteen patients (six males and eight females) were clinically diagnosed as NF1 associated dystrophic scoliosis with common symptoms including café-au-lait spots, paravertebral tumors, and dystrophic scoliosis. NF1 mutations were detected in 12 (85.7%) patients, including four nonsense mutations, three splicing mutations, three frameshift mutations, and two exon deletions. The first surgical procedure included growing-rod surgery in 10 patients and posterior spinal fusion in four patients. The follow-up duration was 2.3 years (1.0-10.3 years), and the Cobb angle of the main curve improved from 61.5° (30°-125°) pre-operatively to 14.5° (0°-42°) at the last follow-up, with an average correction rate of 74.0% (44-100%). Instrumentation-related complications occurred in four patients during the follow-up period. CONCLUSIONS In patients with dystrophic scoliosis who met the clinical diagnostic criteria for NF1, the mutation detection rate of ES combined with MLPA was 85.7%. There was no mutation hotspot in NF1 gene, molecular diagnosis could offer information about genetic counseling, prenatal diagnosis and eugenics. Surgical treatment according to patient's age and severity could effectively correct the spinal deformities.
Collapse
Affiliation(s)
- Haichong Li
- Department of Orthopedics, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Wenyan Zhang
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China.,MOE Key Laboratory of Major Diseases in Children, Beijing, China.,Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.,Henan Key Laboratory of Pediatric Inherited & Metabolic Diseases, Henan Children's Hospital, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Ziming Yao
- Department of Orthopedics, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| | - Ruolan Guo
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China.,MOE Key Laboratory of Major Diseases in Children, Beijing, China.,Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.,Henan Key Laboratory of Pediatric Inherited & Metabolic Diseases, Henan Children's Hospital, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Chanjuan Hao
- Beijing Key Laboratory for Genetics of Birth Defects, Beijing Pediatric Research Institute, Beijing, China.,MOE Key Laboratory of Major Diseases in Children, Beijing, China.,Genetics and Birth Defects Control Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.,Henan Key Laboratory of Pediatric Inherited & Metabolic Diseases, Henan Children's Hospital, Zhengzhou Hospital of Beijing Children's Hospital, Zhengzhou, China
| | - Xuejun Zhang
- Department of Orthopedics, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China
| |
Collapse
|
22
|
de Oliveira Reis L, Fontenele RC, Devito KL, Cunha KS, Domingos ADC. Evaluation of the dimensions, morphology, and position of the mandibular condyles in individuals with neurofibromatosis 1: a case-control study. Clin Oral Investig 2022; 26:159-169. [PMID: 34050829 DOI: 10.1007/s00784-021-03985-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Accepted: 05/10/2021] [Indexed: 12/01/2022]
Abstract
OBJECTIVES The aim of this study was to evaluate the mandibular condyles of neurofibromatosis 1 (NF1) individuals without facial plexiform neurofibroma using cone beam computed tomography images. MATERIALS AND METHODS Eighty cone beam computed tomography scans (160 mandibular condyles) were analyzed: 40 from NF1 individuals (study group) and 40 from individuals without NF1 (control group). The anteroposterior and mediolateral dimensions, height, and volume of the mandibular condyles were measured. The mandibular condyles were classified according to their morphology: healthy (absence of morphological changes), with flattening (loss of rounded contour of at least one of the surfaces), with erosion (loss of continuity of the cortical bone), with osteophyte (exophytic formation of the condyle surface), and with sclerosis (any increase in the cortical thickness in the load-bearing areas). Furthermore, the position of the mandibular condyles in relation to the joint fossa in an anteroposterior view was classified as anterior, concentric, or posterior. RESULTS The study group had a higher anteroposterior dimension of the mandibular condyles compared with the control group (p < 0.05). There were no differences in condylar morphology and position between both groups (p > 0.05). The morphological alterations were not associated with sex or age in any group evaluated (p > 0.05). For both groups, the concentric position was the most common. For the study group, there was a significant difference in the condylar position between the sides (p < 0.05). CONCLUSIONS NF1 individuals without facial plexiform neurofibroma present a high prevalence of condyles with a large anteroposterior dimension and asymmetric position in the joint fossa. However, no morphological and volumetric changes were observed in the mandibular condyles of them. CLINICAL RELEVANCE The knowledge of the TMJ alterations in individuals with NF1 is important to establish an evaluation protocol, which would allow early intervention if indicated.
Collapse
Affiliation(s)
- Larissa de Oliveira Reis
- Department of Oral Diagnosis, Division of Oral Radiology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil.
| | - Rocharles Cavalcante Fontenele
- Department of Oral Diagnosis, Division of Oral Radiology, Piracicaba Dental School, University of Campinas (UNICAMP), Piracicaba, São Paulo, Brazil
| | - Karina Lopes Devito
- Department of Dental Clinic, School of Dentistry, Federal University of Juiz de Fora (UFJF), Juiz de Fora, Minas Gerais, Brazil
| | - Karin Soares Cunha
- Department of Pathology, School of Medicine, Universidade Federal Fluminense (UFF), Niterói, Rio de Janeiro, Brazil
| | - Andréa de Castro Domingos
- Department of Oral Pathology and Diagnosis, School of Dentistry, Federal University of Rio de Janeiro (UFRJ), Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Lv X, Xu J, Jiang J, Wu P, Tan R, Wang B. Genetic animal models of scoliosis: A systematical review. Bone 2021; 152:116075. [PMID: 34174503 DOI: 10.1016/j.bone.2021.116075] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 02/07/2023]
Abstract
Scoliosis is a complex disease with undetermined pathogenesis and has a strong relationship with genetics. Models of scoliosis in animals have been established for better comprehending its pathogenesis and treatment. In this review, we searched all the genetic animal models with body curvature in databases, and reviewed the related genes and scoliosis types. Meanwhile, we also summarized the pathogenesis of scoliosis reported so far. Summarizing the positive phenotypic animal models contributes to a better understanding on the pathogenesis of scoliosis and facilitates the selection of experimental models when a possible pathogenic factor is concerned.
Collapse
Affiliation(s)
- Xin Lv
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jinghong Xu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Jiajiong Jiang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Pengfei Wu
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Renchun Tan
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China
| | - Bing Wang
- Department of Spine Surgery, The Second Xiangya Hospital, Central South University, Changsha 410011, Hunan, China.
| |
Collapse
|
24
|
Fowlkes JL, Thrailkill KM, Bunn RC. RASopathies: The musculoskeletal consequences and their etiology and pathogenesis. Bone 2021; 152:116060. [PMID: 34144233 PMCID: PMC8316423 DOI: 10.1016/j.bone.2021.116060] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 06/07/2021] [Accepted: 06/10/2021] [Indexed: 01/07/2023]
Abstract
The RASopathies comprise an ever-growing number of clinical syndromes resulting from germline mutations in components of the RAS/MAPK signaling pathway. While multiple organs and tissues may be affected by these mutations, this review will focus on how these mutations specifically impact the musculoskeletal system. Herein, we review the genetics and musculoskeletal phenotypes of these syndromes in humans. We discuss how mutations in the RASopathy syndromes have been studied in translational mouse models. Finally, we discuss how signaling molecules within the RAS/MAPK pathway are involved in normal and abnormal bone biology in the context of osteoblasts, osteoclasts and chondrocytes.
Collapse
Affiliation(s)
- John L Fowlkes
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America.
| | - Kathryn M Thrailkill
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| | - R Clay Bunn
- University of Kentucky Barnstable Brown Diabetes Center, Department of Pediatrics, University of Kentucky College of Medicine, Lexington, KY 40536, United States of America
| |
Collapse
|
25
|
Li Y, Zhu M, Lin X, Li J, Yuan Z, Liu Y, Xu H. Autophagy is involved in neurofibromatosis type I gene-modulated osteogenic differentiation in human bone mesenchymal stem cells. Exp Ther Med 2021; 22:1262. [PMID: 34603530 PMCID: PMC8453340 DOI: 10.3892/etm.2021.10697] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 07/14/2021] [Indexed: 12/30/2022] Open
Abstract
Neurofibromatosis type I (NF1) is an autosomal dominant genetic disease that is caused by mutations in the NF1 gene. Various studies have previously demonstrated that the mTOR complex 1 signaling pathway is essential for the NF1-modulated osteogenic differentiation of bone mesenchymal stem cells (BMSCs). Additionally, the mTOR signaling pathway plays a notable role in autophagy. The present study hypothesized that NF1 could modulate the osteogenic differentiation of BMSCs by regulating the autophagic activities of BMSCs. In the present study, human BMSCs were cultured in an osteogenic induction medium. The expression of the NF1 gene was either knocked down or overexpressed by transfection with a specific small interfering RNA (siRNA) targeting NF1 or the pcDNA3.0 NF1-overexpression plasmid, respectively. Autophagic activities of BMSCs (Beclin-1, P62, LC3B I, and LC3B II) were determined using western blotting, electron microscopy, acridine orange (AO) staining and autophagic flux/lysosomal detection by fluorescence microscopy. In addition, the autophagy activator rapamycin (RAPA) and inhibitor 3-methyladenine (3-MA) were used to investigate the effects of autophagy on NF1-modulated osteogenic differentiation in BMSCs. Inhibiting NF1 with siRNA significantly decreased the expression levels of autophagy markers Beclin-1 and LC3B-II, in addition to osteogenic differentiation markers osterix, runt-related transcription factor 2 and alkaline phosphatase. By contrast, overexpressing NF1 with pcDNA3.0 significantly increased their levels. Transmission electron microscopy, AO staining and autophagic flux/lysosomal detection assays revealed that the extent of autophagosome formation was significantly decreased in the NF1-siRNA group but significantly increased in the NF1-pcDNA3.0 group when compared with the NC-siRNA and pcDNA3.0 groups, respectively. In addition, the activity of the PI3K/AKT/mTOR pathway [phosphorylated (p)-PI3K, p-AKT, p-mTOR and p-p70S6 kinase] was significantly upregulated in the NF1-siRNA group compared with the NC-siRNA group, and significantly inhibited in the NF1-pcDNA3.0 group, compared with the pcDNA3.0 group. The knockdown effects of NF1-siRNA on the autophagy and osteogenic differentiation of BMSCs were reversed by the autophagy activator RAPA, while the overexpression effects of NF1-pcDNA3.0 on the autophagy and osteogenic differentiation of BMSCs were reversed by the autophagy inhibitor 3-MA. In conclusion, results from the present study suggest at the involvement of autophagy in the NF1-modulated osteogenic differentiation of BMSCs. Furthermore, NF1 may partially regulate the autophagic activity of BMSCs through the PI3K/AKT/mTOR signaling pathway.
Collapse
Affiliation(s)
- Yiqiang Li
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Mingwei Zhu
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Xuemei Lin
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Jingchun Li
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Zhe Yuan
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Yanhan Liu
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| | - Hongwen Xu
- Department of Pediatric Orthopedics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong 510623, P.R. China
| |
Collapse
|
26
|
Wang W, Wei CJ, Cui XW, Li YH, Gu YH, Gu B, Li QF, Wang ZC. Impacts of NF1 Gene Mutations and Genetic Modifiers in Neurofibromatosis Type 1. Front Neurol 2021; 12:704639. [PMID: 34566848 PMCID: PMC8455870 DOI: 10.3389/fneur.2021.704639] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/15/2021] [Indexed: 12/26/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a tumor predisposition genetic disorder that directly affects more than 1 in 3,000 individuals worldwide. It results from mutations of the NF1 gene and shows almost complete penetrance. NF1 patients show high phenotypic variabilities, including cafe-au-lait macules, freckling, or other neoplastic or non-neoplastic features. Understanding the underlying mechanisms of the diversities of clinical symptoms might contribute to the development of personalized healthcare for NF1 patients. Currently, studies have shown that the different types of mutations in the NF1 gene might correlate with this phenomenon. In addition, genetic modifiers are responsible for the different clinical features. In this review, we summarize different genetic mutations of the NF1 gene and related genetic modifiers. More importantly, we focus on the genotype–phenotype correlation. This review suggests a novel aspect to explain the underlying mechanisms of phenotypic heterogeneity of NF1 and provides suggestions for possible novel therapeutic targets to prevent or delay the onset and development of different manifestations of NF1.
Collapse
Affiliation(s)
- Wei Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Cheng-Jiang Wei
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xi-Wei Cui
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue-Hua Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yi-Hui Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Gu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qing-Feng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Chao Wang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
27
|
Couasnay G, Madel MB, Lim J, Lee B, Elefteriou F. Sites of Cre-recombinase activity in mouse lines targeting skeletal cells. J Bone Miner Res 2021; 36:1661-1679. [PMID: 34278610 DOI: 10.1002/jbmr.4415] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 07/12/2021] [Accepted: 07/15/2021] [Indexed: 12/22/2022]
Abstract
The Cre/Lox system is a powerful tool in the biologist's toolbox, allowing loss-of-function and gain-of-function studies, as well as lineage tracing, through gene recombination in a tissue-specific and inducible manner. Evidence indicates, however, that Cre transgenic lines have a far more nuanced and broader pattern of Cre activity than initially thought, exhibiting "off-target" activity in tissues/cells other than the ones they were originally designed to target. With the goal of facilitating the comparison and selection of optimal Cre lines to be used for the study of gene function, we have summarized in a single manuscript the major sites and timing of Cre activity of the main Cre lines available to target bone mesenchymal stem cells, chondrocytes, osteoblasts, osteocytes, tenocytes, and osteoclasts, along with their reported sites of "off-target" Cre activity. We also discuss characteristics, advantages, and limitations of these Cre lines for users to avoid common risks related to overinterpretation or misinterpretation based on the assumption of strict cell-type specificity or unaccounted effect of the Cre transgene or Cre inducers. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Greig Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
| | | | - Joohyun Lim
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Florent Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
28
|
Zhang T, Han T, Dong Z, Li C, Lu W. Characterization of Two Loss-of-Function NF1 Variants in Chinese Patients and Potential Molecular Interpretations of Phenotypes. Front Genet 2021; 12:660592. [PMID: 34046057 PMCID: PMC8144720 DOI: 10.3389/fgene.2021.660592] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Accepted: 04/13/2021] [Indexed: 11/30/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a common genetic disorder characterized by cafe'-au-lait spots, skinfold freckles, the formation of neurofibromas, skeletal dysplasia, vascular dysplasia, and an increased risk of malignant tumors. In this study, two Chinese NF1 children troubled with bone lesions or hypertension were reported. A de novo NF1 mutation (c.4925T > A/p.V1642E) and a maternally inherited NF1 mutation (c.4883T > A/p.L1628∗) were identified by molecular sequence. According to the ACMG/AMP guidelines, the c.4925T > A was classified as variants of uncertain significance (VOUS) while the c.4883T > A mutation was identified as likely Pathogenic. Further study found that these two NF1 mutants had lost their function to inhibit the Ras/Erk signaling and the proliferation of cells, which could interpretate some phenotypes of these two NF1 patients. We also observed these two NF1 mutants displayed decreased protein stability with increased ubiquitination levels compared with that of wild-type NF1.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Tianting Han
- State Key Laboratory of Molecular Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, Shanghai, China
| | - Zhiya Dong
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| | - Chuanyin Li
- Cancer Center, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Wenli Lu
- Department of Pediatrics, Ruijin Hospital Affiliated to Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
29
|
Russo C, Russo C, Cascone D, Mazio F, Santoro C, Covelli EM, Cinalli G. Non-Oncological Neuroradiological Manifestations in NF1 and Their Clinical Implications. Cancers (Basel) 2021; 13:cancers13081831. [PMID: 33921292 PMCID: PMC8070534 DOI: 10.3390/cancers13081831] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 02/04/2023] Open
Abstract
Simple Summary Central nervous system involvement (CNS) is a common finding in Neurofibromatosis type 1 (NF1). Beside tumor-related manifestations, NF1 is also characterized by a wide spectrum of CNS alterations with variable impacts on functioning and life quality. Here, we propose an overview of non-oncological neuroradiological findings in NF1, with an insight on pathophysiological and embryological clues for a better understanding of the development of these specific alterations. Abstract Neurofibromatosis type 1 (NF1), the most frequent phakomatosis and one of the most common inherited tumor predisposition syndromes, is characterized by several manifestations that pervasively involve central and peripheral nervous system structures. The disorder is due to mutations in the NF1 gene, which encodes for the ubiquitous tumor suppressor protein neurofibromin; neurofibromin is highly expressed in neural crest derived tissues, where it plays a crucial role in regulating cell proliferation, differentiation, and structural organization. This review article aims to provide an overview on NF1 non-neoplastic manifestations of neuroradiological interest, involving both the central nervous system and spine. We also briefly review the most recent MRI functional findings in NF1.
Collapse
Affiliation(s)
- Camilla Russo
- Department of Electrical Engineering and Information Technology (DIETI), University of Naples “Federico II”, 80125 Naples, Italy
- Correspondence: ; Tel.: +39-333-7050711
| | - Carmela Russo
- Pediatric Neuroradiology Unit, Department of Pediatric Neurosciences, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (C.R.); (D.C.); (F.M.); (E.M.C.)
| | - Daniele Cascone
- Pediatric Neuroradiology Unit, Department of Pediatric Neurosciences, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (C.R.); (D.C.); (F.M.); (E.M.C.)
| | - Federica Mazio
- Pediatric Neuroradiology Unit, Department of Pediatric Neurosciences, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (C.R.); (D.C.); (F.M.); (E.M.C.)
| | - Claudia Santoro
- Neurofibromatosis Referral Center, Department of Woman, Child, General and Specialized Surgery, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
- Clinic of Child and Adolescent Neuropsychiatry, Department of Mental and Physical Health, and Preventive Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Eugenio Maria Covelli
- Pediatric Neuroradiology Unit, Department of Pediatric Neurosciences, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy; (C.R.); (D.C.); (F.M.); (E.M.C.)
| | - Giuseppe Cinalli
- Pediatric Neurosurgery Unit, Department of Pediatric Neurosciences, Santobono-Pausilipon Children’s Hospital, 80129 Naples, Italy;
| |
Collapse
|
30
|
Osum SH, Watson AL, Largaespada DA. Spontaneous and Engineered Large Animal Models of Neurofibromatosis Type 1. Int J Mol Sci 2021; 22:1954. [PMID: 33669386 PMCID: PMC7920315 DOI: 10.3390/ijms22041954] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/04/2021] [Indexed: 12/13/2022] Open
Abstract
Animal models are crucial to understanding human disease biology and developing new therapies. By far the most common animal used to investigate prevailing questions about human disease is the mouse. Mouse models are powerful tools for research as their small size, limited lifespan, and defined genetic background allow researchers to easily manipulate their genome and maintain large numbers of animals in general laboratory spaces. However, it is precisely these attributes that make them so different from humans and explains, in part, why these models do not accurately predict drug responses in human patients. This is particularly true of the neurofibromatoses (NFs), a group of genetic diseases that predispose individuals to tumors of the nervous system, the most common of which is Neurofibromatosis type 1 (NF1). Despite years of research, there are still many unanswered questions and few effective treatments for NF1. Genetically engineered mice have drastically improved our understanding of many aspects of NF1, but they do not exemplify the overall complexity of the disease and some findings do not translate well to humans due to differences in body size and physiology. Moreover, NF1 mouse models are heavily reliant on the Cre-Lox system, which does not accurately reflect the molecular mechanism of spontaneous loss of heterozygosity that accompanies human tumor development. Spontaneous and genetically engineered large animal models may provide a valuable supplement to rodent studies for NF1. Naturally occurring comparative models of disease are an attractive prospect because they occur on heterogeneous genetic backgrounds and are due to spontaneous rather than engineered mutations. The use of animals with naturally occurring disease has been effective for studying osteosarcoma, lymphoma, and diabetes. Spontaneous NF-like symptoms including neurofibromas and malignant peripheral nerve sheath tumors (MPNST) have been documented in several large animal species and share biological and clinical similarities with human NF1. These animals could provide additional insight into the complex biology of NF1 and potentially provide a platform for pre-clinical trials. Additionally, genetically engineered porcine models of NF1 have recently been developed and display a variety of clinical features similar to those seen in NF1 patients. Their large size and relatively long lifespan allow for longitudinal imaging studies and evaluation of innovative surgical techniques using human equipment. Greater genetic, anatomic, and physiologic similarities to humans enable the engineering of precise disease alleles found in human patients and make them ideal for preclinical pharmacokinetic and pharmacodynamic studies of small molecule, cellular, and gene therapies prior to clinical trials in patients. Comparative genomic studies between humans and animals with naturally occurring disease, as well as preclinical studies in large animal disease models, may help identify new targets for therapeutic intervention and expedite the translation of new therapies. In this review, we discuss new genetically engineered large animal models of NF1 and cases of spontaneous NF-like manifestations in large animals, with a special emphasis on how these comparative models could act as a crucial translational intermediary between specialized murine models and NF1 patients.
Collapse
Affiliation(s)
- Sara H. Osum
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| | | | - David A. Largaespada
- Masonic Cancer Center, Department of Pediatrics, Division of Hematology and Oncology, University of Minnesota, Minneapolis, MN 55455, USA;
| |
Collapse
|
31
|
Chelleri C, Guerriero V, Torre M, Brolatti N, Piccolo G, Mattioli G, Boero S, Minetti C, Diana MC. Anterior chest wall deformities in children with neurofibromatosis type 1. Acta Paediatr 2021; 110:594-595. [PMID: 32777120 DOI: 10.1111/apa.15529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 08/03/2020] [Accepted: 08/06/2020] [Indexed: 11/26/2022]
Affiliation(s)
- Cristina Chelleri
- Pediatric Neurology and Neuromuscular Disorders Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | - Vittorio Guerriero
- Pediatric Thoracic and Airway Surgery Unit IRCCS Giannina Gaslini Institute Genoa Italy
- Pediatric Surgery Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | - Michele Torre
- Pediatric Thoracic and Airway Surgery Unit IRCCS Giannina Gaslini Institute Genoa Italy
- Pediatric Surgery Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | - Noemi Brolatti
- Pediatric Neurology and Neuromuscular Disorders Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | - Gianluca Piccolo
- Pediatric Neurology and Neuromuscular Disorders Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | | | - Silvio Boero
- Pediatric Orthopedics Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | - Carlo Minetti
- Pediatric Neurology and Neuromuscular Disorders Unit IRCCS Giannina Gaslini Institute Genoa Italy
| | - Maria Cristina Diana
- Pediatric Neurology and Neuromuscular Disorders Unit IRCCS Giannina Gaslini Institute Genoa Italy
| |
Collapse
|
32
|
MEKK2 mediates aberrant ERK activation in neurofibromatosis type I. Nat Commun 2020; 11:5704. [PMID: 33177525 PMCID: PMC7658220 DOI: 10.1038/s41467-020-19555-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 10/14/2020] [Indexed: 12/14/2022] Open
Abstract
Neurofibromatosis type I (NF1) is characterized by prominent skeletal manifestations caused by NF1 loss. While inhibitors of the ERK activating kinases MEK1/2 are promising as a means to treat NF1, the broad blockade of the ERK pathway produced by this strategy is potentially associated with therapy limiting toxicities. Here, we have sought targets offering a more narrow inhibition of ERK activation downstream of NF1 loss in the skeleton, finding that MEKK2 is a novel component of a noncanonical ERK pathway in osteoblasts that mediates aberrant ERK activation after NF1 loss. Accordingly, despite mice with conditional deletion of Nf1 in mature osteoblasts (Nf1fl/fl;Dmp1-Cre) and Mekk2−/− each displaying skeletal defects, Nf1fl/fl;Mekk2−/−;Dmp1-Cre mice show an amelioration of NF1-associated phenotypes. We also provide proof-of-principle that FDA-approved inhibitors with activity against MEKK2 can ameliorate NF1 skeletal pathology. Thus, MEKK2 functions as a MAP3K in the ERK pathway in osteoblasts, offering a potential new therapeutic strategy for the treatment of NF1. Neurofibromatosis type I (NF1) is characterized by prominent skeletal abnormalities mediated in part by aberrant ERK pathway activation due to NF1 loss-of-function. Here, the authors report the MEKK2 is a key mediator of this aberrant ERK activation and that MEKK2 inhibitors, including ponatinib, ameliorate skeletal defects in a mouse model of NF1.
Collapse
|
33
|
Huang S, Jin M, Su N, Chen L. New insights on the reparative cells in bone regeneration and repair. Biol Rev Camb Philos Soc 2020; 96:357-375. [PMID: 33051970 DOI: 10.1111/brv.12659] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022]
Abstract
Bone possesses a remarkable repair capacity to regenerate completely without scar tissue formation. This unique characteristic, expressed during bone development, maintenance and injury (fracture) healing, is performed by the reparative cells including skeletal stem cells (SSCs) and their descendants. However, the identity and functional roles of SSCs remain controversial due to technological difficulties and the heterogeneity and plasticity of SSCs. Moreover, for many years, there has been a biased view that bone marrow is the main cell source for bone repair. Together, these limitations have greatly hampered our understanding of these important cell populations and their potential applications in the treatment of fractures and skeletal diseases. Here, we reanalyse and summarize current understanding of the reparative cells in bone regeneration and repair and outline recent progress in this area, with a particular emphasis on the temporal and spatial process of fracture healing, the sources of reparative cells, an updated definition of SSCs, and markers of skeletal stem/progenitor cells contributing to the repair of craniofacial and long bones, as well as the debate between SSCs and pericytes. Finally, we also discuss the existing problems, emerging novel technologies and future research directions in this field.
Collapse
Affiliation(s)
- Shuo Huang
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Min Jin
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Nan Su
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| | - Lin Chen
- Department of Wound Repair and Rehabilitation Medicine, Center of Bone Metabolism and Repair, State Key Laboratory of Trauma, Burns and Combined Injury, Trauma Center, Research Institute of Surgery, Daping Hospital, Army Medical University (Third Military Medical University), 10 Changjiang zhi Road, Yuzhong District, Chongqing, China
| |
Collapse
|
34
|
Uthoff J, Larson J, Sato TS, Hammond E, Schroeder KE, Rohret F, Rogers CS, Quelle DE, Darbro BW, Khanna R, Weimer JM, Meyerholz DK, Sieren JC. Longitudinal phenotype development in a minipig model of neurofibromatosis type 1. Sci Rep 2020; 10:5046. [PMID: 32193437 PMCID: PMC7081358 DOI: 10.1038/s41598-020-61251-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 02/17/2020] [Indexed: 12/24/2022] Open
Abstract
Neurofibromatosis type 1 (NF1) is a rare, autosomal dominant disease with variable clinical presentations. Large animal models are useful to help dissect molecular mechanisms, determine relevant biomarkers, and develop effective therapeutics. Here, we studied a NF1 minipig model (NF1+/ex42del) for the first 12 months of life to evaluate phenotype development, track disease progression, and provide a comparison to human subjects. Through systematic evaluation, we have shown that compared to littermate controls, the NF1 model develops phenotypic characteristics of human NF1: [1] café-au-lait macules, [2] axillary/inguinal freckling, [3] shortened stature, [4] tibial bone curvature, and [5] neurofibroma. At 4 months, full body computed tomography imaging detected significantly smaller long bones in NF1+/ex42del minipigs compared to controls, indicative of shorter stature. We found quantitative evidence of tibial bowing in a subpopulation of NF1 minipigs. By 8 months, an NF1+/ex42del boar developed a large diffuse shoulder neurofibroma, visualized on magnetic resonance imaging, which subsequently grew in size and depth as the animal aged up to 20 months. The NF1+/ex42del minipig model progressively demonstrates signature attributes that parallel clinical manifestations seen in humans and provides a viable tool for future translational NF1 research.
Collapse
Affiliation(s)
- Johanna Uthoff
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | - Jared Larson
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Takashi S Sato
- Department of Radiology, University of Iowa, Iowa City, IA, USA
| | - Emily Hammond
- Department of Radiology, University of Iowa, Iowa City, IA, USA
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
| | | | | | | | - Dawn E Quelle
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Pharmacology, University of Iowa, Iowa City, IA, USA
| | - Benjamin W Darbro
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA
- Department of Pediatrics, University of Iowa, Iowa City, IA, USA
| | - Rajesh Khanna
- Department of Pharmacology, University of Arizona, Tucson, AZ, USA
| | - Jill M Weimer
- Pediatrics and Rare Diseases Group, Sanford Research, Sioux Falls, SD, USA
| | | | - Jessica C Sieren
- Department of Radiology, University of Iowa, Iowa City, IA, USA.
- Department of Biomedical Engineering, University of Iowa, Iowa City, IA, USA.
- Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA, USA.
| |
Collapse
|
35
|
Prudhomme L, Delleci C, Trimouille A, Chateil JF, Prodhomme O, Goizet C, Van Gils J. Severe Thoracic and Spinal Bone Abnormalities in neurofibromatosis type 1. Eur J Med Genet 2019; 63:103815. [PMID: 31783133 DOI: 10.1016/j.ejmg.2019.103815] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/29/2019] [Accepted: 11/23/2019] [Indexed: 01/15/2023]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant, multi-system, neurocutaneous disorder that predisposes to the development of benign and malignant tumors. Classical skeletal abnormalities encompass sphenoid wing dysplasia, congenital bowing of the long bones and vertebral osteopathy associated with non-dystrophic or dystrophic scoliosis found in about 10% of NF1 patients. We report a 17-year-old boy affected by NF1 with extreme severe spinal and thoracic malformations affecting bone and lung tissues, including hypoplasia of the right lung, unilateral costal agenesis and severe dystrophic scoliosis characterized by association of hemivertebra, fusion of adjacent vertebral bodies and defective pedicles. At birth, he presented an acute respiratory distress requiring invasive ventilator support. The diagnosis of NF1 was confirmed at age 5 by the identification of a de novo heterozygous mutation c.4537C > T, p.Arg1513* in NF1. Trio-based Whole Exome Sequencing (WES) was performed to exclude coexistence of a second hit but no clearly other pathogenic variant has been identified. Until now, only one similar NF1 patient suffering from the same association of severe scoliosis and chest deformity leading to respiratory insufficiency was described. The severe prenatal NF1-related scoliosis could explain the lung abnormal development by absence of mechanical constraints. Severe Thoracic and Spinal Bone Abnormalities may be part of the NF1 bone phenotype and should be taken into account to allow adequate genetic counseling.
Collapse
Affiliation(s)
- L Prudhomme
- Service de Génétique Médicale, CHU Bordeaux, et laboratoire MRGM, INSERM U1211, Univ. Bordeaux, Bordeaux, France
| | - C Delleci
- Centre de Référence Maladies Rares Neurogénétique, Service de Génétique Médicale, CHU, Bordeaux, France; Service de Médecine Physique et de Réadaptation Pôle de Neurosciences Cliniques, CHU de Bordeaux & EA 4136 HACS (handicap Activité Cognition Santé), Université de Bordeaux, France
| | - A Trimouille
- Service de Génétique Médicale, CHU Bordeaux, et laboratoire MRGM, INSERM U1211, Univ. Bordeaux, Bordeaux, France
| | - J F Chateil
- Service d'imagerie antenatale, de l'enfant et de la femme Univ. Bordeaux, RMSB UMR 5536, France
| | - O Prodhomme
- Service d'Imagerie Pédiatrique CHU Hôpital Arnaud de Villeneuve, Montpellier, France
| | - C Goizet
- Service de Génétique Médicale, CHU Bordeaux, et laboratoire MRGM, INSERM U1211, Univ. Bordeaux, Bordeaux, France; Centre de Référence Maladies Rares Neurogénétique, Service de Génétique Médicale, CHU, Bordeaux, France
| | - J Van Gils
- Service de Génétique Médicale, CHU Bordeaux, et laboratoire MRGM, INSERM U1211, Univ. Bordeaux, Bordeaux, France.
| |
Collapse
|
36
|
Zhu G, Zheng Y, Liu Y, Yan A, Hu Z, Yang Y, Xiang S, Li L, Chen W, Peng Y, Zhong N, Mei H. Identification and characterization of NF1 and non-NF1 congenital pseudarthrosis of the tibia based on germline NF1 variants: genetic and clinical analysis of 75 patients. Orphanet J Rare Dis 2019; 14:221. [PMID: 31533797 PMCID: PMC6751843 DOI: 10.1186/s13023-019-1196-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 09/04/2019] [Indexed: 12/13/2022] Open
Abstract
Background Congenital pseudarthrosis of the tibia (CPT) is a rare disease. Some patients present neurofibromatosis type 1 (NF1), while some others do not manifest NF1 (non-NF1). The etiology of CPT, particularly non-NF1 CPT, is not well understood. Here we screened germline variants of 75 CPT cases, including 55 NF1 and 20 non-NF1. Clinical data were classified and analyzed based on NF1 gene variations to investigate the genotype-phenotype relations of the two types of patients. Results Using whole-exome sequencing and Multiplex Ligation-Dependent Probe Amplification, 44 out of 55 NF1 CPT patients (80.0%) were identified as carrying pathogenic variants of the NF1 gene. Twenty-five variants were novel; 53.5% of variants were de novo, and a higher proportion of their carriers presented bone fractures compared to inherited variant carriers. No NF1 pathogenic variants were found in all 20 non-NF1 patients. Clinical features comparing NF1 CPT to non-NF1 CPT did not show significant differences in bowing or fracture onset, lateralization, tissue pathogenical results, abnormality of the proximal tibial epiphysis, and follow-up tibial union after surgery. A considerably higher proportion of non-NF1 patients have cystic lesion (Crawford type III) and used braces after surgery. Conclusions We analyzed a large cohort of non-NF1 and NF1 CPT patients and provided a new perspective for genotype-phenotype features related to germline NF1 variants. Non-NF1 CPT in general had similar clinical features of the tibia as NF1 CPT. Germline NF1 pathogenic variants could differentiate NF1 from non-NF1 CPT but could not explain the CPT heterogeneity of NF1 patients. Our results suggested that non-NF1 CPT was probably not caused by germline NF1 pathogenic variants. In addition to NF1, other genetic variants could also contribute to CPT pathogenesis. Our findings would facilitate the interpretation of NF1 pathogenic variants in CPT genetic counseling. Supplementary information The online version of this article (10.1186/s13023-019-1196-0) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Guanghui Zhu
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The Pediatric Academy of the University of South China, 86# Ziyuan Road, Changsha, Hunan Province, 410007, People's Republic of China
| | - Yu Zheng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China.,Center for Medical Genetics, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, Hunan Province, People's Republic of China
| | - Yaoxi Liu
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The Pediatric Academy of the University of South China, 86# Ziyuan Road, Changsha, Hunan Province, 410007, People's Republic of China
| | - An Yan
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The Pediatric Academy of the University of South China, 86# Ziyuan Road, Changsha, Hunan Province, 410007, People's Republic of China
| | - Zhengmao Hu
- Center for Medical Genetics, School of Life Sciences, Central South University, 110 Xiangya Road, Changsha, Hunan Province, People's Republic of China
| | - Yongjia Yang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China
| | - Shiting Xiang
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China
| | - Liping Li
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China
| | - Weijian Chen
- Pathology Department, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China
| | - Yu Peng
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China
| | - Nanbert Zhong
- Pediatrics Research Institute of Hunan Province, Hunan Children's Hospital, 86 Ziyuan Road, Changsha, Hunan Province, People's Republic of China. .,New York State Institute for Basic Research in Developmental Disabilities, Staten Island, NY, USA.
| | - Haibo Mei
- Department of Pediatric Orthopaedics, Hunan Children's Hospital, The Pediatric Academy of the University of South China, 86# Ziyuan Road, Changsha, Hunan Province, 410007, People's Republic of China.
| |
Collapse
|
37
|
Nissim S, Leshchiner I, Mancias JD, Greenblatt MB, Maertens O, Cassa CA, Rosenfeld JA, Cox AG, Hedgepeth J, Wucherpfennig JI, Kim AJ, Henderson JE, Gonyo P, Brandt A, Lorimer E, Unger B, Prokop JW, Heidel JR, Wang XX, Ukaegbu CI, Jennings BC, Paulo JA, Gableske S, Fierke CA, Getz G, Sunyaev SR, Wade Harper J, Cichowski K, Kimmelman AC, Houvras Y, Syngal S, Williams C, Goessling W. Mutations in RABL3 alter KRAS prenylation and are associated with hereditary pancreatic cancer. Nat Genet 2019; 51:1308-1314. [PMID: 31406347 DOI: 10.1038/s41588-019-0475-y] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 07/01/2019] [Indexed: 12/16/2022]
Abstract
Pancreatic ductal adenocarcinoma is an aggressive cancer with limited treatment options1. Approximately 10% of cases exhibit familial predisposition, but causative genes are not known in most families2. We perform whole-genome sequence analysis in a family with multiple cases of pancreatic ductal adenocarcinoma and identify a germline truncating mutation in the member of the RAS oncogene family-like 3 (RABL3) gene. Heterozygous rabl3 mutant zebrafish show increased susceptibility to cancer formation. Transcriptomic and mass spectrometry approaches implicate RABL3 in RAS pathway regulation and identify an interaction with RAP1GDS1 (SmgGDS), a chaperone regulating prenylation of RAS GTPases3. Indeed, the truncated mutant RABL3 protein accelerates KRAS prenylation and requires RAS proteins to promote cell proliferation. Finally, evidence in patient cohorts with developmental disorders implicates germline RABL3 mutations in RASopathy syndromes. Our studies identify RABL3 mutations as a target for genetic testing in cancer families and uncover a mechanism for dysregulated RAS activity in development and cancer.
Collapse
Affiliation(s)
- Sahar Nissim
- Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA.,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA
| | - Ignaty Leshchiner
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,The Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Joseph D Mancias
- Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Matthew B Greenblatt
- Department of Pathology and Laboratory Medicine and the Hospital for Special Surgery, Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Ophélia Maertens
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Christopher A Cassa
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jill A Rosenfeld
- The Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Andrew G Cox
- Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia.,Department of Biochemistry and Molecular Biology, University of Melbourne, Parkville, Victoria, Australia
| | - John Hedgepeth
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Julia I Wucherpfennig
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Andrew J Kim
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jake E Henderson
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Patrick Gonyo
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Anthony Brandt
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ellen Lorimer
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Bethany Unger
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Jeremy W Prokop
- HudsonAlpha Institute for Biotechnology, Huntsville, AL, USA
| | - Jerry R Heidel
- Carlson College of Veterinary Medicine, Oregon State University, Corvallis, OR, USA
| | | | | | | | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Carol A Fierke
- Department of Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - Gad Getz
- The Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA.,Cancer Center and Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Shamil R Sunyaev
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - J Wade Harper
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Karen Cichowski
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA
| | - Alec C Kimmelman
- Department of Radiation Oncology, Perlmutter Cancer Center, New York University School of Medicine, New York, NY, USA
| | - Yariv Houvras
- Weill Cornell Medical College and New York Presbyterian Hospital, New York, NY, USA
| | - Sapna Syngal
- Gastroenterology Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.,Dana-Farber Cancer Institute, Boston, MA, USA
| | - Carol Williams
- Cancer Center and Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Wolfram Goessling
- Genetics Division, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA. .,Dana-Farber Cancer Institute, Boston, MA, USA. .,Harvard-MIT Division of Health Sciences and Technology, Cambridge, MA, USA. .,The Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, MA, USA. .,Harvard Stem Cell Institute, Cambridge, MA, USA. .,Division of Gastroenterology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
38
|
Korfhage J, Lombard DB. Malignant Peripheral Nerve Sheath Tumors: From Epigenome to Bedside. Mol Cancer Res 2019; 17:1417-1428. [PMID: 31023785 DOI: 10.1158/1541-7786.mcr-19-0147] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 04/08/2019] [Accepted: 04/16/2019] [Indexed: 01/05/2023]
Abstract
Malignant peripheral nerve sheath tumors (MPNST) are aggressive sarcomas typically developing in the context of neurofibromatosis type 1 (NF-1). With the exception of surgical resection, these tumors are resistant to all current therapies, and unresectable, recurrent, or metastatic tumors are considered incurable. Preclinical studies have identified several novel candidate molecular targets for therapeutic intervention, but, to date, targeted therapies have proven ineffective. Recent studies have identified recurrent mutations in polycomb repressive complex 2 (PRC2) core components, embryonic ectoderm development protein (EED) and suppressor of zeste 12 homolog (SUZ12), in MPNST. These mutations result in global loss of the histone H3 lysine 27 trimethylation epigenetic mark, normally deposited by PRC2, and subsequent gain in acetylation at this residue. This altered chromatin state has been shown to promote MPNST malignancy; however, acetylation at this residue sensitizes MPNSTs to BRD4 and bromodomain and extra-terminal domain inhibition. Interestingly, the catalytic component of PRC2, enhancer of zeste homolog 2 (EZH2), is not mutated in MPNST, hinting that a noncanonical, PRC2-independent function of EZH2 may play a role in this cancer. This review examines the pathobiology of MPNST, the contribution of PRC2 subunits to this process, and the prospects for PRC2-related therapies for this cancer. IMPLICATIONS: Identification of mutations in the PRC2 components EED and SUZ12 in the majority of MPNSTs may imply noncanonical oncogenic activities of the intact component, EZH2, and provide new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Justin Korfhage
- Department of Pathology and Institute of Gerontology, University of Michigan, Ann Arbor, Michigan
| | - David B Lombard
- Department of Pathology and Institute of Gerontology, University of Michigan, Ann Arbor, Michigan.
| |
Collapse
|
39
|
Abstract
Scoliosis is a common manifestation of neurofibromatosis type 1, causing significant morbidity. The etiology of dystrophic scoliosis in neurofibromatosis type 1 is not fully understood and therapies are lacking. Somatic mutations in NF1 have been shown in tibial pseudarthrosis providing rationale for similar processes in neurofibromatosis type 1-associated dystrophic scoliosis. Spinal samples from surgical procedures with matched peripheral blood of two individuals with neurofibromatosis type 1 and dystrophic scoliosis were obtained and DNA extracted. Next generation sequencing of various spinal sections as well as the germline/blood sample were performed using a RASopathy gene panel (includes the NF1 gene). Variants were compared between the spinal tissue samples and the germline data. In addition, the next generation sequencing allele frequency data were used to detect somatic loss of heterozygosity. All samples had a detected potentially inactivating NF1 germline mutation. Both individuals demonstrated an allelic imbalance inclusive of NF1 in the next generation sequencing data. In addition, for the same two individuals, there was an increase in the % variant reads for the germline mutation in some of the surgical spinal samples corresponding to the allelic imbalance. Contra analysis did not show any deletion in Chromosome 17 next generation sequencing data. Microarray analysis verified somatic copy neutral loss of heterozygosity for these two individuals for the majority of the chromosome 17 q-arm, inclusive of the NF1 gene. These results suggest that the cause of dystrophic scoliosis is multifactorial and that a somatic NF1 mutation contributes to the etiology.
Collapse
|
40
|
Moro K, Kameyama H, Abe K, Tsuchida J, Tajima Y, Ichikawa H, Nakano M, Ikarashi M, Nagahashi M, Shimada Y, Kato K, Okamoto T, Umezu H, Gabriel E, Tsuchida M, Wakai T. Left colic artery aneurysm rupture after stent placement for abdominal aortic aneurysm associated with neurofibromatosis type 1. Surg Case Rep 2019; 5:12. [PMID: 30673931 PMCID: PMC6346692 DOI: 10.1186/s40792-019-0570-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Accepted: 01/17/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Neurofibromatosis type 1 (NF1) is an autosomal dominant disease of the skin and soft tissue. Aneurysms associated with NF1 can occur, but a secondary aneurysm rupture is very rare, with very few cases reported in literature. CASE PRESENTATION We describe the case of a 67-year-old female with NF1 who underwent endovascular aneurysm repair (EVAR) for an abdominal aortic aneurysm (AAA) rupture. She developed a type Ib endoleak requiring a redo-EVAR. Eighteen days after her primary operation, she was found to have two new left colic artery aneurysms. She required emergency surgery consisting of a left hemicolectomy and transverse colon colostomy. Pathology showed neurofibromatous changes to the peri-vasculature tissue, consistent with her underlying disease. CONCLUSIONS Although rare, secondary aneurysms can occur following AAA repair. Patients with soft tissue connective tissue disorders, like NF1, may be at an increased risk for development of these secondary aneurysms. Endovascular repair appears to be a safe approach for NF1 patients with AAA, but endovascular management can be challenging in the setting of NF1. Surgeons should be ready to convert to open surgery if the patient displays persistent signs of bleeding or structural changes related to connective tissue disorders like NF1.
Collapse
Affiliation(s)
- Kazuki Moro
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hitoshi Kameyama
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan.
| | - Kaoru Abe
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Junko Tsuchida
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yosuke Tajima
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hiroshi Ichikawa
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Masato Nakano
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Mayuko Ikarashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Masayuki Nagahashi
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Yoshifumi Shimada
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Kaori Kato
- Division of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Takeshi Okamoto
- Division of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Hajime Umezu
- Division of Molecular and Diagnostic Pathology, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, 951-8510, Japan
| | | | - Masanori Tsuchida
- Division of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| | - Toshifumi Wakai
- Division of Digestive and General Surgery, Niigata University Graduate School of Medical and Dental Sciences, 1-757 Asahimachi-dori, Chuo-ku, Niigata City, Niigata, 951-8510, Japan
| |
Collapse
|
41
|
Bowden SA, Foster BL. Alkaline Phosphatase Replacement Therapy for Hypophosphatasia in Development and Practice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2019; 1148:279-322. [PMID: 31482504 DOI: 10.1007/978-981-13-7709-9_13] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hypophosphatasia (HPP) is an inherited disorder that affects bone and tooth mineralization characterized by low serum alkaline phosphatase. HPP is caused by loss-of-function mutations in the ALPL gene encoding the protein, tissue-nonspecific alkaline phosphatase (TNSALP). TNSALP is expressed by mineralizing cells of the skeleton and dentition and is associated with the mineralization process. Generalized reduction of activity of the TNSALP leads to accumulation of its substrates, including inorganic pyrophosphate (PPi) that inhibits physiological mineralization. This leads to defective skeletal mineralization, with manifestations including rickets, osteomalacia, fractures, and bone pain, all of which can result in multi-systemic complications with significant morbidity, as well as mortality in severe cases. Dental manifestations are nearly universal among affected individuals and feature most prominently premature loss of deciduous teeth. Management of HPP has been limited to supportive care until the introduction of a TNSALP enzyme replacement therapy (ERT), asfotase alfa (AA). AA ERT has proven to be transformative, improving survival in severely affected infants and increasing overall quality of life in children and adults with HPP. This chapter provides an overview of TNSALP expression and functions, summarizes HPP clinical types and pathologies, discusses early attempts at therapies for HPP, summarizes development of HPP mouse models, reviews design and validation of AA ERT, and provides up-to-date accounts of AA ERT efficacy in clinical trials and case reports, including therapeutic response, adverse effects, limitations, and potential future directions in therapy.
Collapse
Affiliation(s)
- S A Bowden
- Division of Endocrinology, Department of Pediatrics, Nationwide Children's Hospital/The Ohio State University College of Medicine, Columbus, OH, USA.
| | - B L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
42
|
Lu J, Zhu LF, Cai YM, Dong HY, Zhu L, Tan JM. Isolation and multipotential differentiation of mesenchymal stromal cell‑like progenitor cells from human bladder. Mol Med Rep 2018; 19:187-194. [PMID: 30431114 PMCID: PMC6297775 DOI: 10.3892/mmr.2018.9646] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 10/19/2018] [Indexed: 02/07/2023] Open
Abstract
Various types of mesenchymal stromal cells (MSCs) have been used in urological tissue engineering but to date the existence of MSCs has not been reported in the human bladder. The present study provided evidence that a small number of MSC‑like cells exist in the human bladder and designated this class of cells 'human bladder‑derived MSC‑like cells' (hBSCs). It was demonstrated that hBSCs can be cultured to yield a large population. These hBSCs expressed the surface markers of MSCs and exhibited the capacity for osteogenic, adipogenic and chondrogenic differentiation. On induction with appropriate media in vitro, hBSCs could differentiate into bladder‑associated cell types, including urothelial, endothelial and smooth muscle cell‑like lineages. In addition, the average telomerase activity of adult hBSCs was higher compared with adult human bone marrow‑derived MSCs, but lower than that of human umbilical cord Wharton's jelly‑derived MSCs. These findings may inspire future studies on the role of hBSCs in urological tissue engineering applications and in other fields.
Collapse
Affiliation(s)
- Jun Lu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Ling-Feng Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Yuan-Ming Cai
- College of Basic Medical, Xinjiang Medical University, Urumqi, Xinjiang Uygur Autonomous Region 830001, P.R. China
| | - Hui-Yue Dong
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Ling Zhu
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| | - Jian-Ming Tan
- Fujian Provincial Key Laboratory of Transplant Biology, Fuzhou General Hospital/ or Dongfang Hospital, Xiamen University, Fuzhou, Fujian 350025, P.R. China
| |
Collapse
|
43
|
Deo N, El-Hoss J, Kolind M, Mikulec K, Peacock L, Little DG, Schindeler A. JNK inhibitor CC-930 reduces fibrosis in a murine model of Nf1-deficient fracture repair. J Appl Biomed 2018. [DOI: 10.1016/j.jab.2018.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
|
44
|
Tajan M, Paccoud R, Branka S, Edouard T, Yart A. The RASopathy Family: Consequences of Germline Activation of the RAS/MAPK Pathway. Endocr Rev 2018; 39:676-700. [PMID: 29924299 DOI: 10.1210/er.2017-00232] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 06/13/2018] [Indexed: 12/13/2022]
Abstract
Noonan syndrome [NS; Mendelian Inheritance in Men (MIM) #163950] and related syndromes [Noonan syndrome with multiple lentigines (formerly called LEOPARD syndrome; MIM #151100), Noonan-like syndrome with loose anagen hair (MIM #607721), Costello syndrome (MIM #218040), cardio-facio-cutaneous syndrome (MIM #115150), type I neurofibromatosis (MIM #162200), and Legius syndrome (MIM #611431)] are a group of related genetic disorders associated with distinctive facial features, cardiopathies, growth and skeletal abnormalities, developmental delay/mental retardation, and tumor predisposition. NS was clinically described more than 50 years ago, and disease genes have been identified throughout the last 3 decades, providing a molecular basis to better understand their physiopathology and identify targets for therapeutic strategies. Most of these genes encode proteins belonging to or regulating the so-called RAS/MAPK signaling pathway, so these syndromes have been gathered under the name RASopathies. In this review, we provide a clinical overview of RASopathies and an update on their genetics. We then focus on the functional and pathophysiological effects of RASopathy-causing mutations and discuss therapeutic perspectives and future directions.
Collapse
Affiliation(s)
- Mylène Tajan
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Romain Paccoud
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Sophie Branka
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| | - Thomas Edouard
- Endocrine, Bone Diseases, and Genetics Unit, Children's Hospital, Toulouse University Hospital, Toulouse, France
| | - Armelle Yart
- INSERM UMR 1048, Institute of Cardiovascular and Metabolic Diseases (I2MC), University of Toulouse Paul Sabatier, Toulouse, France
| |
Collapse
|
45
|
Almeida PN, Barboza DDN, Luna EB, Correia MCDM, Dias RB, Siquara de Sousa AC, Duarte MEL, Rossi MID, Cunha KS. Increased extracellular matrix deposition during chondrogenic differentiation of dental pulp stem cells from individuals with neurofibromatosis type 1: an in vitro 2D and 3D study. Orphanet J Rare Dis 2018; 13:98. [PMID: 29941005 PMCID: PMC6020206 DOI: 10.1186/s13023-018-0843-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 06/14/2018] [Indexed: 12/12/2022] Open
Abstract
Background Neurofibromatosis 1 (NF1) presents a wide range of clinical manifestations, including bone alterations. Studies that seek to understand cellular and molecular mechanisms underlying NF1 orthopedic problems are of great importance to better understand the pathogenesis and the development of new therapies. Dental pulp stem cells (DPSCs) are being used as an in vitro model for several diseases and appear as a suitable model for NF1. The aim of this study was to evaluate in vitro chondrogenic differentiation of DPSCs from individuals with NF1 using two-dimensional (2D) and three-dimensional (3D) cultures. Results To fulfill the criteria of the International Society for Cellular Therapy, DPSCs were characterized by surface antigen expression and by their multipotentiality, being induced to differentiate towards adipogenic, osteogenic, and chondrogenic lineages in 2D cultures. Both DPSCs from individuals with NF1 (NF1 DPSCs) and control cultures were positive for CD90, CD105, CD146 and negative for CD13, CD14, CD45 and CD271, and successfully differentiated after the protocols. Chondrogenic differentiation was evaluated in 2D and in 3D (pellet) cultures, which were further evaluated by optical microscopy and transmission electron microscopy (TEM). 2D cultures showed greater extracellular matrix deposition in NF1 DPSCs comparing with controls during chondrogenic differentiation. In semithin sections, control pellets hadhomogenous-sized intra and extracelullar matrix vesicles, whereas NF1 cultures had matrix vesicles of different sizes. TEM analysis showed higher amount of collagen fibers in NF1 cultures compared with control cultures. Conclusion NF1 DPSCs presented increased extracellular matrix deposition during chondrogenic differentiation, which could be related to skeletal changes in individuals with NF1. Electronic supplementary material The online version of this article (10.1186/s13023-018-0843-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Paula Nascimento Almeida
- Graduate Program in Pathology, School of Medicine, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil.,Neurofibromatosis National Center (Centro Nacional de Neurofibromatose), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Deuilton do Nascimento Barboza
- Oral and Maxillofacial Surgery, Antônio Pedro University Hospital, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil
| | - Eloá Borges Luna
- Graduate Program in Pathology, School of Medicine, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil.,Neurofibromatosis National Center (Centro Nacional de Neurofibromatose), Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Rhayra Braga Dias
- National Institute of Traumatology and Orthopedics (Instituto Nacional de Traumatologia e Ortopedia), Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Maria Eugenia Leite Duarte
- National Institute of Traumatology and Orthopedics (Instituto Nacional de Traumatologia e Ortopedia), Rio de Janeiro, Rio de Janeiro, Brazil
| | - Maria Isabel Doria Rossi
- Institute of Biomedical Sciences, and Clementino Fraga Filho University Hospital, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Karin Soares Cunha
- Graduate Program in Pathology, School of Medicine, Universidade Federal Fluminense, Niterói, Rio de Janeiro, Brazil. .,Neurofibromatosis National Center (Centro Nacional de Neurofibromatose), Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
46
|
Luna EB, Janini MER, Lima F, Pontes RRA, Guedes FR, Geller M, da Silva LE, Motta AT, Cunha KS. Craniomaxillofacial morphology alterations in children, adolescents and adults with neurofibromatosis 1: A cone beam computed tomography analysis of a Brazilian sample. Med Oral Patol Oral Cir Bucal 2018; 23:e168-e179. [PMID: 29476678 PMCID: PMC5911357 DOI: 10.4317/medoral.22155] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 09/05/2017] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Oral manifestations are common in neurofibromatosis 1 (NF1), and include jaws and teeth alterations. Our aim was to investigate the craniomaxillofacial morphology of Brazilian children, adolescents and adults with NF1 using cone beam computed tomography. MATERIAL AND METHODS This study was conducted with 36 Brazilian individuals with NF1 with ages ranging from 4 to 75. The participants were submitted to anamnesis, extra and intraoral exam and cephalometric analysis using cone beam computed tomography. Height of the NF1 individuals was compared to the length of jaws and skull base. The results of the cephalometric measurements of the NF1 group were compared with a control group paired by age, gender and skin color. RESULTS Individuals with NF1 had lower maxillary length (p<0.0001), lower mandibular length (p<0.0001), lower skull base length (p<0.0001). In children and adolescents, the mandible was more posteriorly positioned (p=0.01), when compared with the control group. There was no association between jaws and skull base length with the height of the individuals with NF1. CONCLUSIONS Brazilian children, adolescents and adults with NF1 have short mandible, maxilla and skull base. Moreover, children and adolescents present mandibular retrusion.
Collapse
Affiliation(s)
- E-B Luna
- School of Medicine, Hospital Universitário Antônio Pedro, Universidade Federal Fluminense, Av. Marquês do Paraná, 303, 4o andar, sala 01, Centro, Niterói, RJ, Brazil, Zip code: 24033-900,
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Patra D, DeLassus E, Mueller J, Abou-Ezzi G, Sandell LJ. Site-1 protease regulates skeletal stem cell population and osteogenic differentiation in mice. Biol Open 2018; 7:bio.032094. [PMID: 29437042 PMCID: PMC5861364 DOI: 10.1242/bio.032094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Site-1 protease (S1P) is a proprotein convertase with essential functions in the conversion of precursor proteins to their active form. In earlier studies, we demonstrated that S1P ablation in the chondrocyte lineage results in a drastic reduction in endochondral bone formation. To investigate the mechanistic contribution of S1P to bone development we ablated S1P in the osterix lineage in mice. S1P ablation in this lineage results in osteochondrodysplasia and variable degrees of early postnatal scoliosis. Embryonically, even though Runx2 and osterix expression are normal, S1P ablation results in a delay in vascular invasion and endochondral bone development. Mice appear normal when born, but by day 7 display pronounced dwarfism with fragile bones that exhibit significantly reduced mineral density, mineral apposition rate, bone formation rate and reduced osteoblasts indicating severe osteopenia. Mice suffer from a drastic reduction in bone marrow mesenchymal progenitors as analyzed by colony-forming unit-fibroblast assay. Fluorescence-activated cell sorting analysis of the skeletal mesenchyme harvested from bone marrow and collagenase-digested bone show a drastic reduction in hematopoietic lineage-negative, endothelial-negative, CD105+ skeletal stem cells. Bone marrow mesenchymal progenitors are unable to differentiate into osteoblasts in vitro, with no effect on adipogenic differentiation. Postnatal mice have smaller growth plates with reduced hypertrophic zone. Thus, S1P controls bone development directly by regulating the skeletal progenitor population and their differentiation into osteoblasts. This article has an associated First Person interview with the first author of the paper. Summary: S1P governs a fundamental aspect of skeletal development and homeostasis, mainly the maintenance and osteogenic differentiation of skeletogenic stem cells that are a source of osteoblast and chondrocyte lineages.
Collapse
Affiliation(s)
- Debabrata Patra
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Elizabeth DeLassus
- Department of Biochemistry, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jennifer Mueller
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Grazia Abou-Ezzi
- Department of Medicine, Oncology Division, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Linda J Sandell
- Department of Orthopaedic Surgery, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| |
Collapse
|
48
|
Tahaei SE, Couasnay G, Ma Y, Paria N, Gu J, Lemoine BF, Wang X, Rios JJ, Elefteriou F. The reduced osteogenic potential of Nf1-deficient osteoprogenitors is EGFR-independent. Bone 2018; 106:103-111. [PMID: 29032173 PMCID: PMC5694354 DOI: 10.1016/j.bone.2017.10.012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/28/2017] [Accepted: 10/10/2017] [Indexed: 12/26/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common genetic disorder caused by mutations in the NF1 gene. Recalcitrant bone healing following fracture (i.e. pseudarthrosis) is one of the most problematic skeletal complications associated with NF1. The etiology of this condition is still unclear; thus, pharmacological options for clinical management are limited. Multiple studies have shown the reduced osteogenic potential of Nf1-deficient osteoprogenitors. A recent transcriptome profiling investigation revealed that EREG and EGFR, encoding epiregulin and its receptor Epidermal Growth Factor Receptor 1, respectively, were among the top over-expressed genes in cells of the NF1 pseudarthrosis site. Because EGFR stimulation is known to inhibit osteogenic differentiation, we hypothesized that increased EREG and EGFR expression in NF1-deficient skeletal progenitors may contribute to their reduced osteogenic differentiation potential. In this study, we first confirmed via single-cell mRNA sequencing that EREG over-expression was associated with NF1 second hit somatic mutations in human bone cells, whereas Transforming Growth Factor beta 1 (TGFβ1) expression was unchanged. Second, using ex-vivo recombined Nf1-deficient mouse bone marrow stromal cells (mBMSCs), we show that this molecular signature is conserved between mice and humans, and that epiregulin generated by these cells is overexpressed and active, whereas soluble TGFβ1 expression and activity are not affected. However, blocking either epiregulin function or EGFR signaling by EGFR1 or pan EGFR inhibition (using AG-1478 and Poziotinib respectively) did not correct the differentiation defect of Nf1-deficient mBMSCs, as measured by the expression of Alpl, Ibsp and alkaline phosphatase activity. These results suggest that clinically available drugs aimed at inhibiting EGFR signaling are unlikely to have a significant benefit for the management of bone non-union in children with NF1 PA.
Collapse
Affiliation(s)
- S E Tahaei
- Department of Pharmacology, Vanderbilt University, Nashville, TN, United States; Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - G Couasnay
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - Y Ma
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States
| | - N Paria
- Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, United States
| | - J Gu
- Baylor Institute for Immunology Research, Dallas, TX, United States
| | - B F Lemoine
- Baylor Institute for Immunology Research, Dallas, TX, United States
| | - X Wang
- Baylor Institute for Immunology Research, Dallas, TX, United States
| | - J J Rios
- Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, United States; Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, United States; McDermott Center for Human Growth and Development, UT Southwestern Medical Center, Dallas, TX, United States; Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, TX, United States
| | - F Elefteriou
- Department of Orthopedic Surgery, Baylor College of Medicine, Houston, TX, United States; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, United States.
| |
Collapse
|
49
|
Newey PJ, Thakker RV. Multiple Endocrine Neoplasia Syndromes. GENETICS OF BONE BIOLOGY AND SKELETAL DISEASE 2018:699-732. [DOI: 10.1016/b978-0-12-804182-6.00038-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
50
|
Summers MA, Rupasinghe T, Vasiljevski ER, Evesson FJ, Mikulec K, Peacock L, Quinlan KGR, Cooper ST, Roessner U, Stevenson DA, Little DG, Schindeler A. Dietary intervention rescues myopathy associated with neurofibromatosis type 1. Hum Mol Genet 2017; 27:577-588. [DOI: 10.1093/hmg/ddx423] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2017] [Accepted: 11/29/2017] [Indexed: 02/07/2023] Open
Affiliation(s)
- Matthew A Summers
- Orthopaedic Research & Biotechnology, The Children’s Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Paediatrics & Child Heath, Faculty of Medicine, University of Sydney, Camperdown, NSW, Australia
| | | | - Emily R Vasiljevski
- Orthopaedic Research & Biotechnology, The Children’s Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Paediatrics & Child Heath, Faculty of Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Frances J Evesson
- Institute for Neuroscience and Muscle Research, The Children’s Hospital Westmead, Sydney, NSW, Australia
| | - Kathy Mikulec
- Orthopaedic Research & Biotechnology, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Lauren Peacock
- Orthopaedic Research & Biotechnology, The Children’s Hospital at Westmead, Westmead, NSW, Australia
| | - Kate G R Quinlan
- Discipline of Paediatrics & Child Heath, Faculty of Medicine, University of Sydney, Camperdown, NSW, Australia
- Institute for Neuroscience and Muscle Research, The Children’s Hospital Westmead, Sydney, NSW, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, NSW, Australia
| | - Sandra T Cooper
- Discipline of Paediatrics & Child Heath, Faculty of Medicine, University of Sydney, Camperdown, NSW, Australia
- Institute for Neuroscience and Muscle Research, The Children’s Hospital Westmead, Sydney, NSW, Australia
| | - Ute Roessner
- Metabolomics Australia, University of Melbourne, VIC, Australia
| | - David A Stevenson
- Division of Medical Genetics, Stanford University, Stanford, CA, USA
| | - David G Little
- Orthopaedic Research & Biotechnology, The Children’s Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Paediatrics & Child Heath, Faculty of Medicine, University of Sydney, Camperdown, NSW, Australia
| | - Aaron Schindeler
- Orthopaedic Research & Biotechnology, The Children’s Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Paediatrics & Child Heath, Faculty of Medicine, University of Sydney, Camperdown, NSW, Australia
| |
Collapse
|