51
|
Monroe CL, Dahiya S, Gutmann DH. Dissecting Clinical Heterogeneity in Neurofibromatosis Type 1. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2017; 12:53-74. [PMID: 28135565 DOI: 10.1146/annurev-pathol-052016-100228] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Neurofibromatosis type 1 (NF1) is a common neurogenetic disorder in which affected children and adults are predisposed to the development of benign and malignant nervous system tumors. Caused by a germline mutation in the NF1 tumor suppressor gene, individuals with NF1 are prone to optic gliomas, malignant gliomas, neurofibromas, and malignant peripheral nerve sheath tumors, as well as behavioral, cognitive, motor, bone, cardiac, and pigmentary abnormalities. Although NF1 is a classic monogenic syndrome, the clinical features of the disorder and their impact on patient morbidity are variable, even within individuals who bear the same germline NF1 gene mutation. As such, NF1 affords unique opportunities to define the factors that contribute to disease heterogeneity and to develop therapies personalized to a given individual (precision medicine). This review highlights the clinical features of NF1 and the use of genetically engineered mouse models to define the molecular and cellular pathogenesis of NF1-associated nervous system tumors.
Collapse
Affiliation(s)
- Courtney L Monroe
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110;
| | - Sonika Dahiya
- Division of Neuropathology, Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, Missouri 63110
| | - David H Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri 63110;
| |
Collapse
|
52
|
Kamiya N, Yamaguchi R, Aruwajoye O, Kim AJ, Kuroyanagi G, Phipps M, Adapala NS, Feng JQ, Kim HK. Targeted Disruption of NF1 in Osteocytes Increases FGF23 and Osteoid With Osteomalacia-like Bone Phenotype. J Bone Miner Res 2017; 32:1716-1726. [PMID: 28425622 DOI: 10.1002/jbmr.3155] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Revised: 04/11/2017] [Accepted: 04/16/2017] [Indexed: 12/30/2022]
Abstract
Neurofibromatosis type 1 (NF1, OMIM 162200), caused by NF1 gene mutations, exhibits multi-system abnormalities, including skeletal deformities in humans. Osteocytes play critical roles in controlling bone modeling and remodeling. However, the role of neurofibromin, the protein product of the NF1 gene, in osteocytes is largely unknown. This study investigated the role of neurofibromin in osteocytes by disrupting Nf1 under the Dmp1-promoter. The conditional knockout (Nf1 cKO) mice displayed serum profile of a metabolic bone disorder with an osteomalacia-like bone phenotype. Serum FGF23 levels were 4 times increased in cKO mice compared with age-matched controls. In addition, calcium-phosphorus metabolism was significantly altered (calcium reduced; phosphorus reduced; parathyroid hormone [PTH] increased; 1,25(OH)2 D decreased). Bone histomorphometry showed dramatically increased osteoid parameters, including osteoid volume, surface, and thickness. Dynamic bone histomorphometry revealed reduced bone formation rate and mineral apposition rate in the cKO mice. TRAP staining showed a reduced osteoclast number. Micro-CT demonstrated thinner and porous cortical bones in the cKO mice, in which osteocyte dendrites were disorganized as assessed by electron microscopy. Interestingly, the cKO mice exhibited spontaneous fractures in long bones, as found in NF1 patients. Mechanical testing of femora revealed significantly reduced maximum force and stiffness. Immunohistochemistry showed significantly increased FGF23 protein in the cKO bones. Moreover, primary osteocytes from cKO femora showed about eightfold increase in FGF23 mRNA levels compared with control cells. The upregulation of FGF23 was specifically and significantly inhibited by PI3K inhibitor Ly294002, indicating upregulation of FGF23 through PI3K in Nf1-deficient osteocytes. Taken together, these results indicate that Nf1 deficiency in osteocytes dramatically increases FGF23 production and causes a mineralization defect (ie, hyperosteoidosis) via the alteration of calcium-phosphorus metabolism. This study demonstrates critical roles of neurofibromin in osteocytes for osteoid mineralization. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nobuhiro Kamiya
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA.,Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA.,Sports Medicine, Tenri University, Tenri, Japan
| | | | | | - Audrey J Kim
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Gen Kuroyanagi
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Matthew Phipps
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | - Naga Suresh Adapala
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA.,Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jian Q Feng
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, USA
| | - Harry Kw Kim
- Texas Scottish Rite Hospital for Children, Dallas, TX, USA.,Orthopaedic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
53
|
Franceschi RT, Ge C. Control of the Osteoblast Lineage by Mitogen-Activated Protein Kinase Signaling. ACTA ACUST UNITED AC 2017; 3:122-132. [PMID: 29057206 DOI: 10.1007/s40610-017-0059-5] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
PURPOSE OF THE REVIEW This review will provide a timely assessment of MAP kinase actions in bone development and homeostasis with particular emphasis on transcriptional control of the osteoblast lineage. RECENT FINDINGS ERK and p38 MAP kinases function as transducers of signals initiated by the extracellular matrix, mechanical loading, TGF-β, BMPs and FGF2. MAPK signals may also affect and/or interact with other important pathways such as WNT and HIPPO. ERK and p38 MAP kinase pathways phosphorylate specific osteogenic transcription factors including RUNX2, Osterix, ATF4 and DLX5. For RUNX2, phosphorylation at specific serine residues initiates epigenetic changes in chromatin necessary for decondensation and increased transcription. MAPK also suppresses marrow adipogenesis by phosphorylating and inhibiting PPARγ, which may explain the well-known relationship between reduced skeletal loading and marrow fat accumulation. SUMMARY MAPKs transduce signals from the extracellular environment to the nucleus allowing bone cells to respond to changes in hormonal/growth factor signaling and mechanical loading thereby optimizing bone structure to meet physiological and mechanical needs of the body.
Collapse
Affiliation(s)
- Renny T Franceschi
- Departments of Periodontics and Oral Medicine, University of Michigan School of Dentistry and Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109-1078
| | - Chunxi Ge
- Departments of Periodontics and Oral Medicine, University of Michigan School of Dentistry and Department of Biological Chemistry, University of Michigan School of Medicine, Ann Arbor, MI 48109-1078
| |
Collapse
|
54
|
Abstract
Neurofibromatosis type 1 is a complex autosomal dominant disorder caused by germline mutations in the NF1 tumour suppressor gene. Nearly all individuals with neurofibromatosis type 1 develop pigmentary lesions (café-au-lait macules, skinfold freckling and Lisch nodules) and dermal neurofibromas. Some individuals develop skeletal abnormalities (scoliosis, tibial pseudarthrosis and orbital dysplasia), brain tumours (optic pathway gliomas and glioblastoma), peripheral nerve tumours (spinal neurofibromas, plexiform neurofibromas and malignant peripheral nerve sheath tumours), learning disabilities, attention deficits, and social and behavioural problems, which can negatively affect quality of life. With the identification of NF1 and the generation of accurate preclinical mouse strains that model some of these clinical features, therapies that target the underlying molecular and cellular pathophysiology for neurofibromatosis type 1 are becoming available. Although no single treatment exists, current clinical management strategies include early detection of disease phenotypes (risk assessment) and biologically targeted therapies. Similarly, new medical and behavioural interventions are emerging to improve the quality of life of patients. Although considerable progress has been made in understanding this condition, numerous challenges remain; a collaborative and interdisciplinary approach is required to manage individuals with neurofibromatosis type1 and to develop effective treatments.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University School of Medicine, Box 8111, 660 S. Euclid Avenue, St. Louis, Missouri 63110, USA
| | - Rosalie E Ferner
- Department of Neurology, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- Department of Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Robert H Listernick
- Department of Academic General Pediatrics and Primary Care, Ann and Robert H. Lurie Children's Hospital of Chicago, Chicago, Illinois, USA
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Bruce R Korf
- Department of Genetics, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Pamela L Wolters
- Pediatric Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | | |
Collapse
|
55
|
Abstract
The RAS/MAPK signaling pathway plays key roles in development, cell survival and proliferation, as well as in cancer pathogenesis. Molecular genetic studies have identified a group of developmental syndromes, the RASopathies, caused by germ line mutations in this pathway. The syndromes included within this classification are neurofibromatosis type 1 (NF1), Noonan syndrome (NS), Noonan syndrome with multiple lentigines (NS-ML, formerly known as LEOPARD syndrome), Costello syndrome (CS), cardio-facio-cutaneous syndrome (CFC), Legius syndrome (LS, NF1-like syndrome), capillary malformation-arteriovenous malformation syndrome (CM-AVM), and hereditary gingival fibromatosis (HGF) type 1. Although these syndromes present specific molecular alterations, they are characterized by a large spectrum of functional and morphological abnormalities, which include heart defects, short stature, neurocognitive impairment, craniofacial malformations, and, in some cases, cancer predisposition. The development of genetically modified animals, such as mice (Mus musculus), flies (Drosophila melanogaster), and zebrafish (Danio rerio), has been instrumental in elucidating the molecular and cellular bases of these syndromes. Moreover, these models can also be used to determine tumor predisposition, the impact of different genetic backgrounds on the variable phenotypes found among the patients and to evaluate preventative and therapeutic strategies. Here, we review a wide range of genetically modified mouse models used in the study of RASopathies and the potential application of novel technologies, which hopefully will help us resolve open questions in the field.
Collapse
|
56
|
Cnbp ameliorates Treacher Collins Syndrome craniofacial anomalies through a pathway that involves redox-responsive genes. Cell Death Dis 2016; 7:e2397. [PMID: 27711076 PMCID: PMC5133970 DOI: 10.1038/cddis.2016.299] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2016] [Revised: 07/28/2016] [Accepted: 08/25/2016] [Indexed: 11/08/2022]
Abstract
Treacher Collins Syndrome (TCS) is a rare congenital disease (1:50 000 live births) characterized by craniofacial defects, including hypoplasia of facial bones, cleft palate and palpebral fissures. Over 90% of the cases are due to mutations in the TCOF1 gene, which codifies the nucleolar protein Treacle. Here we report a novel TCS-like zebrafish model displaying features that fully recapitulate the spectrum of craniofacial abnormalities observed in patients. As it was reported for a Tcof1+/- mouse model, Treacle depletion in zebrafish caused reduced rRNA transcription, stabilization of Tp53 and increased cell death in the cephalic region. An increase of ROS along with the overexpression of redox-responsive genes was detected; furthermore, treatment with antioxidants ameliorated the phenotypic defects of craniofacial anomalies in TCS-like larvae. On the other hand, Treacle depletion led to a lowering in the abundance of Cnbp, a protein required for proper craniofacial development. Tcof1 knockdown in transgenic zebrafish overexpressing cnbp resulted in barely affected craniofacial cartilage development, reinforcing the notion that Cnbp has a role in the pathogenesis of TCS. The cnbp overexpression rescued the TCS phenotype in a dose-dependent manner by a ROS-cytoprotective action that prevented the redox-responsive genes' upregulation but did not normalize the synthesis of rRNAs. Finally, a positive correlation between the expression of CNBP and TCOF1 in mesenchymal cells from both control and TCS subjects was found. Based on this, we suggest CNBP as an additional target for new alternative therapeutic treatments to reduce craniofacial defects not only in TCS but also in other neurocristopathies.
Collapse
|
57
|
Jindal GA, Goyal Y, Burdine RD, Rauen KA, Shvartsman SY. RASopathies: unraveling mechanisms with animal models. Dis Model Mech 2016. [PMID: 26203125 PMCID: PMC4527292 DOI: 10.1242/dmm.020339] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
RASopathies are developmental disorders caused by germline mutations in the Ras-MAPK pathway, and are characterized by a broad spectrum of functional and morphological abnormalities. The high incidence of these disorders (∼1/1000 births) motivates the development of systematic approaches for their efficient diagnosis and potential treatment. Recent advances in genome sequencing have greatly facilitated the genotyping and discovery of mutations in affected individuals, but establishing the causal relationships between molecules and disease phenotypes is non-trivial and presents both technical and conceptual challenges. Here, we discuss how these challenges could be addressed using genetically modified model organisms that have been instrumental in delineating the Ras-MAPK pathway and its roles during development. Focusing on studies in mice, zebrafish and Drosophila, we provide an up-to-date review of animal models of RASopathies at the molecular and functional level. We also discuss how increasingly sophisticated techniques of genetic engineering can be used to rigorously connect changes in specific components of the Ras-MAPK pathway with observed functional and morphological phenotypes. Establishing these connections is essential for advancing our understanding of RASopathies and for devising rational strategies for their management and treatment. Summary: Developmental disorders caused by germline mutations in the Ras-MAPK pathway are called RASopathies. Studies with animal models, including mice, zebrafish and Drosophila, continue to enhance our understanding of these diseases.
Collapse
Affiliation(s)
- Granton A Jindal
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Yogesh Goyal
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Rebecca D Burdine
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Katherine A Rauen
- Department of Pediatrics, MIND Institute, Division of Genomic Medicine, University of California, Davis, Sacramento, CA 95817, USA
| | - Stanislav Y Shvartsman
- Department of Chemical and Biological Engineering, Princeton University, Princeton, NJ 08544, USA Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
58
|
Activation of the Extracellular Signal-Regulated Kinase Signaling Is Critical for Human Umbilical Cord Mesenchymal Stem Cell Osteogenic Differentiation. BIOMED RESEARCH INTERNATIONAL 2016; 2016:3764372. [PMID: 26989682 PMCID: PMC4771893 DOI: 10.1155/2016/3764372] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 01/15/2016] [Accepted: 01/21/2016] [Indexed: 01/12/2023]
Abstract
Human umbilical cord mesenchymal stem cells (hUCMSCs) are recognized as candidate progenitor cells for bone regeneration. However, the mechanism of hUCMSC osteogenesis remains unclear. In this study, we revealed that mitogen-activated protein kinases (MAPKs) signaling is involved in hUCMSC osteogenic differentiation in vitro. Particularly, the activation of c-Jun N-terminal kinases (JNK) and p38 signaling pathways maintained a consistent level in hUCMSCs through the entire 21-day osteogenic differentiation period. At the same time, the activation of extracellular signal-regulated kinases (ERK) signaling significantly increased from day 5, peaked at day 9, and declined thereafter. Moreover, gene profiling of osteogenic markers, alkaline phosphatase (ALP) activity measurement, and alizarin red staining demonstrated that the application of U0126, a specific inhibitor for ERK activation, completely prohibited hUCMSC osteogenic differentiation. However, when U0126 was removed from the culture at day 9, ERK activation and osteogenic differentiation of hUCMSCs were partially recovered. Together, these findings demonstrate that the activation of ERK signaling is essential for hUCMSC osteogenic differentiation, which points out the significance of ERK signaling pathway to regulate the osteogenic differentiation of hUCMSCs as an alternative cell source for bone tissue engineering.
Collapse
|
59
|
Bakker AC, La Rosa S, Sherman LS, Knight P, Lee H, Pancza P, Nievo M. Neurofibromatosis as a gateway to better treatment for a variety of malignancies. Prog Neurobiol 2016; 152:149-165. [PMID: 26854064 DOI: 10.1016/j.pneurobio.2016.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 01/25/2016] [Accepted: 01/25/2016] [Indexed: 12/23/2022]
Abstract
The neurofibromatoses (NF) are a group of rare genetic disorders that can affect all races equally at an incidence from 1:3000 (NF1) to a log unit lower for NF2 and schwannomatosis. Since the research community is reporting an increasing number of malignant cancers that carry mutations in the NF genes, the general interest of both the research and pharma community is increasing and the authors saw an opportunity to present a novel, fresh approach to drug discovery in NF. The aim of the paper is to challenge the current drug discovery approach to NF, whereby existing targeted therapies that are either in the clinic or on the market for other disease indications are repurposed for NF. We offer a suggestion for an alternative drug discovery approach. In the new approach, selective and tolerable targeted therapies would be developed for NF and later expanded to patients with more complex diseases such as malignant cancer in which the NF downstream pathways are deregulated. The Children's Tumor Foundation, together with some other major NF funders, is playing a key role in funding critical initiatives that will accelerate the development of better targeted therapies for NF patients, while these novel, innovative treatments could potentially be beneficial to molecularly characterized cancer patients in which NF mutations have been identified.
Collapse
Affiliation(s)
- Annette C Bakker
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Salvatore La Rosa
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Larry S Sherman
- Division of Neuroscience, Oregon National Primate Research Center, Oregon Health & Science University, 505 NW 185th Avenue, Beaverton, OR 97006, United States
| | - Pamela Knight
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Hyerim Lee
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Patrice Pancza
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States
| | - Marco Nievo
- Children's Tumor Foundation, 120, Wall Street, 16th Floor, New York 10005, United States.
| |
Collapse
|
60
|
Rhodes SD, Yang FC. Aberrant Myeloid Differentiation Contributes to the Development of Osteoporosis in Neurofibromatosis Type 1. Curr Osteoporos Rep 2016; 14:10-5. [PMID: 26932441 DOI: 10.1007/s11914-016-0298-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurofibromatosis type 1 (NF1), also known as von Recklinghausen disease, is a common autosomal dominant genetic disorder affecting approximately 1 in 3000 individuals worldwide. NF1 results from heritable or spontaneous mutations of the NF1 tumor suppressor gene. NF1 encodes the protein neurofibromin, which functions to negatively regulate Ras-activity. Approximately 50 % of NF1 patients develop osteopenia or osteoporosis, resulting in significantly increased rates of long-bone fracture and morbidity. While defective osteoblast bone anabolism has been implicated as a central factor in the pathogenesis of NF1 associated skeletal deficits, recent data suggest that NF1 (Nf1) haploinsufficiency within the hematopoietic compartment, particularly in osteoclasts and myeloid progenitors, plays a pivotal role in engendering NF1 osseous manifestations. In this chapter, we review the latest data from clinical studies and murine models delineating a critical role for hematopoietic compartment, myeloid progenitors of NF1 (Nf1) haploinsufficient and their progeny-osteoclasts, in the pathogenesis of NF1 associated osteopenia/osteoporosis and discuss putative targets for future therapeutics.
Collapse
Affiliation(s)
- Steven D Rhodes
- Department of Pediatrics, Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Feng-Chun Yang
- Sylvester Comprehensive Cancer Center, Department of Biochemistry and Molecular Biology, University of Miami School of Medicine, R. Bunn Gautier Building, 417 1011 NW 15th street, Locator R-629, Miami, FL, 33136, USA.
| |
Collapse
|
61
|
Zhong ZA, Zahatnansky J, Snider J, Van Wieren E, Diegel CR, Williams BO. Wntless spatially regulates bone development through β-catenin-dependent and independent mechanisms. Dev Dyn 2015; 244:1347-55. [PMID: 26249818 DOI: 10.1002/dvdy.24316] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2015] [Revised: 06/30/2015] [Accepted: 07/06/2015] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Canonical and noncanonical Wnt signaling pathways both play pivotal roles in bone development. Wntless/GPR177 is a chaperone protein that is required for secretion of all Wnt ligands. We previously showed that deletion of Wntless within mature osteoblasts severely impaired postnatal bone homeostasis. RESULTS In this study, we systemically evaluated how deletion of Wntless in different stages of osteochondral differentiation affected embryonic bone development, by crossing Wntless (Wls)-flox/flox mice with strains expressing cre recombinase behind the following promoters: Osteocalcin, Collagen 2a1, or Dermo1. Ex vivo µCT and whole-mount skeletal staining were performed to examine skeletal mineralization. Histology and immunohistochemistry were used to evaluate cellular differentiation and alterations in Wnt signaling. In this work, we found that Wntless regulated chondrogenesis and osteogenesis through both canonical and noncanonical Wnt signaling. CONCLUSIONS These findings provide more insight into the requirements of different Wnt-secretion cell types critical for skeletal development.
Collapse
Affiliation(s)
- Zhendong A Zhong
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Juraj Zahatnansky
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - John Snider
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Emily Van Wieren
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Cassandra R Diegel
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| | - Bart O Williams
- Program for Skeletal Disease and Tumor Microenvironment and Center for Cancer and Cell Biology, Van Andel Research Institute, Grand Rapids, Michigan
| |
Collapse
|
62
|
Abstract
Mesenchymal progenitors of the osteogenic lineage provide the flexibility for bone to grow, maintain its function and homeostasis. Traditionally, colony-forming-unit fibroblasts (CFU-Fs) have been regarded as surrogates for mesenchymal progenitors; however, this definition cannot address the function of these progenitors in their native setting. Transgenic murine models including lineage-tracing technologies based on the cre-lox system have proven to be useful in delineating mesenchymal progenitors in their native environment. Although heterogeneity of cell populations of interest marked by a promoter-based approach complicates overall interpretation, an emerging complexity of mesenchymal progenitors has been revealed. Current literatures suggest two distinct types of bone progenitor cells; growth-associated mesenchymal progenitors contribute to explosive growth of bone in early life, whereas bone marrow mesenchymal progenitors contribute to the much slower remodeling process and response to injury that occurs mainly in adulthood. More detailed relationships of these progenitors need to be studied through further experimentation.
Collapse
Affiliation(s)
- Noriaki Ono
- Department of Orthodontics and Pediatric Dentistry, University of Michigan School of Dentistry, Ann Arbor, MI, 48109, USA
| | - Henry M Kronenberg
- Endocrine Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA,
| |
Collapse
|
63
|
Abstract
Fibroblast growth factor (FGF) signaling pathways are essential regulators of vertebrate skeletal development. FGF signaling regulates development of the limb bud and formation of the mesenchymal condensation and has key roles in regulating chondrogenesis, osteogenesis, and bone and mineral homeostasis. This review updates our review on FGFs in skeletal development published in Genes & Development in 2002, examines progress made on understanding the functions of the FGF signaling pathway during critical stages of skeletogenesis, and explores the mechanisms by which mutations in FGF signaling molecules cause skeletal malformations in humans. Links between FGF signaling pathways and other interacting pathways that are critical for skeletal development and could be exploited to treat genetic diseases and repair bone are also explored.
Collapse
Affiliation(s)
- David M Ornitz
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, Missouri 63110, USA
| | - Pierre J Marie
- UMR-1132, Institut National de la Santé et de la Recherche Médicale, Hopital Lariboisiere, 75475 Paris Cedex 10, France; Université Paris Diderot, Sorbonne Paris Cité, 75475 Paris Cedex 10, France
| |
Collapse
|
64
|
Lu L, Harutyunyan K, Jin W, Wu J, Yang T, Chen Y, Joeng KS, Bae Y, Tao J, Dawson BC, Jiang MM, Lee B, Wang LL. RECQL4 Regulates p53 Function In Vivo During Skeletogenesis. J Bone Miner Res 2015; 30:1077-89. [PMID: 25556649 DOI: 10.1002/jbmr.2436] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 11/30/2014] [Accepted: 12/12/2014] [Indexed: 11/05/2022]
Abstract
RECQ DNA helicases play critical roles in maintaining genomic stability, but their role in development has been less well studied. Rothmund-Thomson syndrome, RAPADILINO, and Baller-Gerold syndrome are rare genetic disorders caused by mutations in the RECQL4 gene. These patients have significant skeletal developmental abnormalities including radial ray, limb and craniofacial defects. To investigate the role of Recql4 in the developing skeletal system, we generated Recql4 conditional knockout mice targeting the skeletal lineage. Inactivation of Recql4 using the Prx1-Cre transgene led to limb abnormalities and craniosynostosis mimicking the major bone findings in human RECQL4 patients. These Prx1-Cre(+) ;Recql4(fl/fl) mice as well as Col2a1-Cre(+) ;Recql4(fl/fl) mice exhibited growth plate defects and an increased p53 response in affected tissues. Inactivation of Trp53 in these Recql4 mutants resulted in genetic rescue of the skeletal phenotypes, indicating an in vivo interaction between Recql4 and Trp53, and p53 activation as an underlying mechanism for the developmental bone abnormalities in RECQL4 disorders. Our findings show that RECQL4 is critical for skeletal development by modulating p53 activity in vivo.
Collapse
Affiliation(s)
- Linchao Lu
- Texas Children's Cancer Center, Department of Pediatrics, Houston, TX, USA
| | - Karine Harutyunyan
- Texas Children's Cancer Center, Department of Pediatrics, Houston, TX, USA
| | - Weidong Jin
- Texas Children's Cancer Center, Department of Pediatrics, Houston, TX, USA
| | - Jianhong Wu
- Texas Children's Cancer Center, Department of Pediatrics, Houston, TX, USA
| | - Tao Yang
- Center for Skeletal Diseases and Tumor Metastasis, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Yuqing Chen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Howard Hughes Medical Institute, Houston, TX, USA
| | - Kyu Sang Joeng
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Yangjin Bae
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Jianning Tao
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brian C Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Howard Hughes Medical Institute, Houston, TX, USA
| | - Ming-Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Howard Hughes Medical Institute, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.,Howard Hughes Medical Institute, Houston, TX, USA
| | - Lisa L Wang
- Texas Children's Cancer Center, Department of Pediatrics, Houston, TX, USA
| |
Collapse
|
65
|
Karner CM, Long F, Solnica-Krezel L, Monk KR, Gray RS. Gpr126/Adgrg6 deletion in cartilage models idiopathic scoliosis and pectus excavatum in mice. Hum Mol Genet 2015; 24:4365-73. [PMID: 25954032 DOI: 10.1093/hmg/ddv170] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/05/2015] [Indexed: 01/27/2023] Open
Abstract
Adolescent idiopathic scoliosis (AIS) and pectus excavatum (PE) are common pediatric musculoskeletal disorders. Little is known about the tissue of origin for either condition, or about their genetic bases. Common variants near GPR126/ADGRG6 (encoding the adhesion G protein-coupled receptor 126/adhesion G protein-coupled receptor G6, hereafter referred to as GPR126) were recently shown to be associated with AIS in humans. Here, we provide genetic evidence that loss of Gpr126 in osteochondroprogenitor cells alters cartilage biology and spinal column development. Microtomographic and x-ray studies revealed several hallmarks of AIS, including postnatal onset of scoliosis without malformations of vertebral units. The mutants also displayed a dorsal-ward deflection of the sternum akin to human PE. At the cellular level, these defects were accompanied by failure of midline fusion within the developing annulus fibrosis of the intervertebral discs and increased apoptosis of chondrocytes in the ribs and vertebrae. Molecularly, we found that loss of Gpr126 upregulated the expression of Gal3st4, a gene implicated in human PE, encoding Galactose-3-O-sulfotransferase 4. Together, these data uncover Gpr126 as a genetic cause for the pathogenesis of AIS and PE in a mouse model.
Collapse
Affiliation(s)
| | - Fanxin Long
- Department of Orthopaedic Surgery, Department of Medicine and Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Lilianna Solnica-Krezel
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Kelly R Monk
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - Ryan S Gray
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO 63110, USA
| |
Collapse
|
66
|
Bobyn JD, Little DG, Gray R, Schindeler A. Animal models of scoliosis. J Orthop Res 2015; 33:458-67. [PMID: 25492698 DOI: 10.1002/jor.22797] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 12/03/2014] [Indexed: 02/04/2023]
Abstract
Multiple techniques designed to induce scoliotic deformity have been applied across many animal species. We have undertaken a review of the literature regarding experimental models of scoliosis in animals to discuss their utility in comprehending disease aetiology and treatment. Models of scoliosis in animals can be broadly divided into quadrupedal and bipedal experiments. Quadrupedal models, in the absence of axial gravitation force, depend upon development of a mechanical asymmetry along the spine to initiate a scoliotic deformity. Bipedal models more accurately mimic human posture and consequently are subject to similar forces due to gravity, which have been long appreciated to be a contributing factor to the development of scoliosis. Many effective models of scoliosis in smaller animals have not been successfully translated to primates and humans. Though these models may not clarify the aetiology of human scoliosis, by providing a reliable and reproducible deformity in the spine they are a useful means with which to test interventions designed to correct and prevent deformity.
Collapse
Affiliation(s)
- Justin D Bobyn
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Sydney, Australia; Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, Sydney, Australia
| | | | | | | |
Collapse
|
67
|
Rhodes SD, Zhang W, Yang D, Yang H, Chen S, Wu X, Li X, Yang X, Mohammad KS, Guise TA, Bergner AL, Stevenson DA, Yang FC. Dystrophic spinal deformities in a neurofibromatosis type 1 murine model. PLoS One 2015; 10:e0119093. [PMID: 25786243 PMCID: PMC4364663 DOI: 10.1371/journal.pone.0119093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2014] [Accepted: 01/16/2015] [Indexed: 12/28/2022] Open
Abstract
Despite the high prevalence and significant morbidity of spinal anomalies in neurofibromatosis type 1 (NF1), the pathogenesis of these defects remains largely unknown. Here, we present two murine models: Nf1flox/−;PeriCre and Nf1flox/−;Col.2.3Cre mice, which recapitulate spinal deformities seen in the human disease. Dynamic histomorphometry and microtomographic studies show recalcitrant bone remodeling and distorted bone microarchitecture within the vertebral spine of Nf1flox/−;PeriCre and Nf1flox/−;Col2.3Cre mice, with analogous histological features present in a human patient with dystrophic scoliosis. Intriguingly, 36–60% of Nf1flox/−;PeriCre and Nf1flox/−;Col2.3Cre mice exhibit segmental vertebral fusion anomalies with boney obliteration of the intervertebral disc (IVD). While analogous findings have not yet been reported in the NF1 patient population, we herein present two case reports of IVD defects and interarticular vertebral fusion in patients with NF1. Collectively, these data provide novel insights regarding the pathophysiology of dystrophic spinal anomalies in NF1, and provide impetus for future radiographic analyses of larger patient cohorts to determine whether IVD and vertebral fusion defects may have been previously overlooked or underreported in the NF1 patient population.
Collapse
Affiliation(s)
- Steven D. Rhodes
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Wei Zhang
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Hebei Medical University, The Third Hospital, Shijiazhuang, China
| | - Dalong Yang
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Hebei Medical University, The Third Hospital, Shijiazhuang, China
| | - Hao Yang
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Shi Chen
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xiaohua Wu
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xiaohong Li
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Xianlin Yang
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Khalid S. Mohammad
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Theresa A. Guise
- Department of Endocrinology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
| | - Amanda L. Bergner
- Department of Neurology, Johns Hopkins Hospital, Baltimore, Maryland, United States of America
| | - David A. Stevenson
- Division of Medical Genetics, Department of Pediatrics, Stanford University, Stanford, California, United States of America
| | - Feng-Chun Yang
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, United States of America
- * E-mail:
| |
Collapse
|
68
|
Karolak MR, Yang X, Elefteriou F. FGFR1 signaling in hypertrophic chondrocytes is attenuated by the Ras-GAP neurofibromin during endochondral bone formation. Hum Mol Genet 2015; 24:2552-64. [PMID: 25616962 DOI: 10.1093/hmg/ddv019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Accepted: 01/20/2015] [Indexed: 01/22/2023] Open
Abstract
Aberrant fibroblast growth factor receptor 3 (FGFR3) signaling disrupts chondrocyte proliferation and growth plate size and architecture, leading to various chondrodysplasias or bone overgrowth. These observations suggest that the duration, intensity and cellular context of FGFR signaling during growth plate chondrocyte maturation require tight, regulated control for proper bone elongation. However, the machinery fine-tuning FGFR signaling in chondrocytes is incompletely defined. We report here that neurofibromin, a Ras-GAP encoded by Nf1, has an overlapping expression pattern with FGFR1 and FGFR3 in prehypertrophic chondrocytes, and with FGFR1 in hypertrophic chondrocytes during endochondral ossification. Based on previous evidence that neurofibromin inhibits Ras-ERK signaling in chondrocytes and phenotypic analogies between mice with constitutive FGFR1 activation and Nf1 deficiency in Col2a1-positive chondrocytes, we asked whether neurofibromin is required to control FGFR1-Ras-ERK signaling in maturing chondrocytes in vivo. Genetic Nf1 ablation in Fgfr1-deficient chondrocytes reactivated Ras-ERK1/2 signaling in hypertrophic chondrocytes and reversed the expansion of the hypertrophic zone observed in mice lacking Fgfr1 in Col2a1-positive chondrocytes. Histomorphometric and gene expression analyses suggested that neurofibromin, by inhibiting Rankl expression, attenuates pro-osteoclastogenic FGFR1 signaling in hypertrophic chondrocytes. We also provide evidence suggesting that neurofibromin in prehypertrophic chondrocytes, downstream of FGFRs and via an indirect mechanism, is required for normal extension and organization of proliferative columns. Collectively, this study indicates that FGFR signaling provides an important input into the Ras-Raf-MEK-ERK1/2 signaling axis in chondrocytes, and that this input is differentially regulated during chondrocyte maturation by a complex intracellular machinery, of which neurofibromin is a critical component.
Collapse
Affiliation(s)
| | - Xiangli Yang
- Department of Pharmacology, Vanderbilt Center for Bone Biology, Department of Medicine and
| | - Florent Elefteriou
- Department of Pharmacology, Vanderbilt Center for Bone Biology, Department of Medicine and Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
69
|
|
70
|
de la Croix Ndong J, Stevens DM, Vignaux G, Uppuganti S, Perrien DS, Yang X, Nyman JS, Harth E, Elefteriou F. Combined MEK inhibition and BMP2 treatment promotes osteoblast differentiation and bone healing in Nf1Osx -/- mice. J Bone Miner Res 2015; 30:55-63. [PMID: 25043591 PMCID: PMC4280331 DOI: 10.1002/jbmr.2316] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2014] [Revised: 06/19/2014] [Accepted: 07/08/2014] [Indexed: 12/24/2022]
Abstract
Neurofibromatosis type I (NF1) is an autosomal dominant disease with an incidence of 1/3000, caused by mutations in the NF1 gene, which encodes the RAS/GTPase-activating protein neurofibromin. Non-bone union after fracture (pseudarthrosis) in children with NF1 remains a challenging orthopedic condition to treat. Recent progress in understanding the biology of neurofibromin suggested that NF1 pseudarthrosis stems primarily from defects in the bone mesenchymal lineage and hypersensitivity of hematopoietic cells to TGFβ. However, clinically relevant pharmacological approaches to augment bone union in these patients remain limited. In this study, we report the generation of a novel conditional mutant mouse line used to model NF1 pseudoarthrosis, in which Nf1 can be ablated in an inducible fashion in osteoprogenitors of postnatal mice, thus circumventing the dwarfism associated with previous mouse models where Nf1 is ablated in embryonic mesenchymal cell lineages. An ex vivo-based cell culture approach based on the use of Nf1(flox/flox) bone marrow stromal cells showed that loss of Nf1 impairs osteoprogenitor cell differentiation in a cell-autonomous manner, independent of developmental growth plate-derived or paracrine/hormonal influences. In addition, in vitro gene expression and differentiation assays indicated that chronic ERK activation in Nf1-deficient osteoprogenitors blunts the pro-osteogenic property of BMP2, based on the observation that only combination treatment with BMP2 and MEK inhibition promoted the differentiation of Nf1-deficient osteoprogenitors. The in vivo preclinical relevance of these findings was confirmed by the improved bone healing and callus strength observed in Nf1osx (-/-) mice receiving Trametinib (a MEK inhibitor) and BMP2 released locally at the fracture site via a novel nanoparticle and polyglycidol-based delivery method. Collectively, these results provide novel evidence for a cell-autonomous role of neurofibromin in osteoprogenitor cells and insights about a novel targeted approach for the treatment of NF1 pseudoarthrosis.
Collapse
Affiliation(s)
- Jean de la Croix Ndong
- Vanderbilt Center for Bone Biology
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - David M. Stevens
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Guillaume Vignaux
- Vanderbilt Center for Bone Biology
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Sasidhar Uppuganti
- Vanderbilt Center for Bone Biology
- Department of Orthopaedic Surgery & Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Daniel S. Perrien
- Vanderbilt Center for Bone Biology
- Department of Orthopaedic Surgery & Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
- Vanderbilt University Institute of Imaging Sciences, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Xiangli Yang
- Vanderbilt Center for Bone Biology
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Jeffry S. Nyman
- Vanderbilt Center for Bone Biology
- Department of Orthopaedic Surgery & Rehabilitation, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, USA
| | - Eva Harth
- Department of Chemistry, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | - Florent Elefteriou
- Vanderbilt Center for Bone Biology
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| |
Collapse
|
71
|
Proposing the use of dental pulp stem cells as a suitable biological model of neurofibromatosis type 1. Childs Nerv Syst 2015; 31:7-13. [PMID: 25480698 DOI: 10.1007/s00381-014-2599-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2014] [Accepted: 11/24/2014] [Indexed: 01/09/2023]
Abstract
PURPOSE This study aims to propose the dental pulp stem cells (DPSCs) as a model for studying two features related to neurofibromatosis type 1 (NF1), i.e. augmented proliferative capacity and altered osteogenic differentiation. METHODS We isolated a DPSC from the pulp of deciduous teeth of a 6-year-old NF1 patient and two other healthy children of similar age. Cell proliferation was assayed by counting with a haemocytometer after successive cell re-plating. In order to compare osteogenic differentiation, we used osteoblast-differentiating medium and quantified alizarin stain, which relates to degree of calcification, and evaluated the expression of osteoblastic markers by reverse transcription polymerase chain reaction (RT-PCR). RESULTS The DPSCs isolated from the NF1 patient displayed a greater rate of proliferation when compared to the control cells. Osteogenic differentiation occurred as expected for both NF1 and control, which concerned cell morphology and expression of osteoblast marker genes ALP, BMP2, BMP4, OCN and SPP1. However, alizarin staining denoted a markedly lower calcification level in the cells from the NF1-diagnosed child, considering that less calcium deposits were visualized under light microscopy and a smaller amount of alizarin could be quantified by spectrophotometry after extraction from the stained cells. CONCLUSION DPSCs seem to be useful as a model for studying NF1 and predicting prognosis of patients, since their in vitro behaviour seems to mimic at least two features of this disorder: higher tendency to develop bone abnormalities and neoplastic cell proliferation.
Collapse
|
72
|
Genetic and functional studies of the intervertebral disc: a novel murine intervertebral disc model. PLoS One 2014; 9:e112454. [PMID: 25474689 PMCID: PMC4256369 DOI: 10.1371/journal.pone.0112454] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2014] [Accepted: 10/06/2014] [Indexed: 12/21/2022] Open
Abstract
Intervertebral disc (IVD) homeostasis is mediated through a combination of micro-environmental and biomechanical factors, all of which are subject to genetic influences. The aim of this study is to develop and characterize a genetically tractable, ex vivo organ culture model that can be used to further elucidate mechanisms of intervertebral disc disease. Specifically, we demonstrate that IVD disc explants (1) maintain their native phenotype in prolonged culture, (2) are responsive to exogenous stimuli, and (3) that relevant homeostatic regulatory mechanisms can be modulated through ex-vivo genetic recombination. We present a novel technique for isolation of murine IVD explants with demonstration of explant viability (CMFDA/propidium iodide staining), disc anatomy (H&E), maintenance of extracellular matrix (ECM) (Alcian Blue staining), and native expression profile (qRT-PCR) as well as ex vivo genetic recombination (mT/mG reporter mice; AdCre) following 14 days of culture in DMEM media containing 10% fetal bovine serum, 1% L-glutamine, and 1% penicillin/streptomycin. IVD explants maintained their micro-anatomic integrity, ECM proteoglycan content, viability, and gene expression profile consistent with a homeostatic drive in culture. Treatment of genetically engineered explants with cre-expressing adenovirus efficaciously induced ex vivo genetic recombination in a variety of genetically engineered mouse models. Exogenous administration of IL-1ß and TGF-ß3 resulted in predicted catabolic and anabolic responses, respectively. Genetic recombination of TGFBR1fl/fl explants resulted in constitutively active TGF-ß signaling that matched that of exogenously administered TGF-ß3. Our results illustrate the utility of the murine intervertebral disc explant to investigate mechanisms of intervertebral disc degeneration.
Collapse
|
73
|
Paria N, Cho TJ, Choi IH, Kamiya N, Kayembe K, Mao R, Margraf RL, Obermosser G, Oxendine I, Sant DW, Song MH, Stevenson DA, Viskochil DH, Wise CA, Kim HKW, Rios JJ. Neurofibromin deficiency-associated transcriptional dysregulation suggests a novel therapy for tibial pseudoarthrosis in NF1. J Bone Miner Res 2014; 29:2636-42. [PMID: 24932921 PMCID: PMC4268180 DOI: 10.1002/jbmr.2298] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Revised: 06/09/2014] [Accepted: 06/12/2014] [Indexed: 12/25/2022]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant disease caused by mutations in NF1. Among the earliest manifestations is tibial pseudoarthrosis and persistent nonunion after fracture. To further understand the pathogenesis of pseudoarthrosis and the underlying bone remodeling defect, pseudoarthrosis tissue and cells cultured from surgically resected pseudoarthrosis tissue from NF1 individuals were analyzed using whole-exome and whole-transcriptome sequencing as well as genomewide microarray analysis. Genomewide analysis identified multiple genetic mechanisms resulting in somatic biallelic NF1 inactivation; no other genes with recurring somatic mutations were identified. Gene expression profiling identified dysregulated pathways associated with neurofibromin deficiency, including phosphoinositide 3-kinase (PI3K) and mitogen-activated protein kinase (MAPK) signaling pathways. Unlike aggressive NF1-associated malignancies, tibial pseudoarthrosis tissue does not harbor a high frequency of somatic mutations in oncogenes or other tumor-suppressor genes, such as p53. However, gene expression profiling indicates that pseudoarthrosis tissue has a tumor-promoting transcriptional pattern, despite lacking tumorigenic somatic mutations. Significant overexpression of specific cancer-associated genes in pseudoarthrosis highlights a potential for receptor tyrosine kinase inhibitors to target neurofibromin-deficient pseudoarthrosis and promote proper bone remodeling and fracture healing.
Collapse
Affiliation(s)
- Nandina Paria
- Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, TX, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
A subset of chondrogenic cells provides early mesenchymal progenitors in growing bones. Nat Cell Biol 2014; 16:1157-67. [PMID: 25419849 PMCID: PMC4250334 DOI: 10.1038/ncb3067] [Citation(s) in RCA: 322] [Impact Index Per Article: 29.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/16/2014] [Indexed: 12/31/2022]
Abstract
The hallmark of endochondral bone development is the presence of cartilaginous templates, in which osteoblasts and stromal cells are generated to form mineralized matrix and support bone marrow hematopoiesis. However, the ultimate source of these mesenchymal cells and the relationship between bone progenitors in fetal life and those in later life are unknown. Fate-mapping studies revealed that cells expressing cre-recombinases driven by the collagen II (Col2) promoter/enhancer and their descendants contributed to, in addition to chondrocytes, early perichondrial precursors prior to Runx2 expression and, subsequently, to a majority of osteoblasts, Cxcl12 (chemokine (C-X-C motif) ligand 12)-abundant stromal cells and bone marrow stromal/mesenchymal progenitor cells in postnatal life. Lineage-tracing experiments using a tamoxifen-inducible creER system further revealed that early postnatal cells marked by Col2-creER, as well as Sox9-creER and aggrecan (Acan)-creER, progressively contributed to multiple mesenchymal lineages and continued to provide descendants for over a year. These cells are distinct from adult mesenchymal progenitors and thus provide opportunities for regulating the explosive growth that occurs uniquely in growing mammals.
Collapse
|
75
|
El-Hoss J, Cheng T, Carpenter EC, Sullivan K, Deo N, Mikulec K, Little DG, Schindeler A. A Combination of rhBMP-2 (Recombinant Human Bone Morphogenetic Protein-2) and MEK (MAP Kinase/ERK Kinase) Inhibitor PD0325901 Increases Bone Formation in a Murine Model of Neurofibromatosis Type I Pseudarthrosis. J Bone Joint Surg Am 2014; 96:e117. [PMID: 25031379 DOI: 10.2106/jbjs.m.00862] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Congenital tibial dysplasia is a severe pediatric condition that classically results in a persistent pseudarthrosis. A majority of these cases are associated with neurofibromatosis type I (NF1), a genetic disorder in which inactivation of the NF1 gene leads to overactivity of the Ras-MEK-MAPK (mitogen-activated protein kinase) signaling pathway. We therefore hypothesized that pharmaceutical inhibition of MEK-MAPK may be a beneficial therapeutic strategy. METHODS In vitro methods were used to demonstrate a role for the MEK inhibitor PD0325901 in promoting osteogenic differentiation in Nf1-/- calvarial osteoblasts. Local applications of rhBMP-2 and/or PD0325901 were then tested in a mouse model of NF1 tibial pseudarthrosis featuring localized double inactivation of the Nf1 gene in a fracture. Mice received no treatment, PD0325901 (10 mg/kg/day from two days before fracture to ten days after fracture), rhBMP-2 (10 μg), or a combination of rhBMP-2 and PD0325901. RESULTS Animals treated with the delivery vehicle alone, PD0325901, rhBMP-2, or the PD0325901 + rhBMP-2 combination showed union rates of 0%, 8%, 69% (p < 0.01), or 80% (p < 0.01), respectively, at twenty-one days after fracture. Mice treated with the rhBMP-2 + PD0325901 combination displayed a callus volume sixfold greater than the vehicle controls and twofold greater than the group receiving rhBMP-2 alone. Although MEK inhibition combined with rhBMP-2 led to increases in bone formation and union, the proportion of fibrous tissue in the callus was not significantly reduced. CONCLUSIONS The data suggest that MEK inhibition can promote bone formation in combination with rhBMP-2 in the context of an NF1 pseudarthrosis. However, PD0325901 did not promote substantive bone anabolism in the absence of an exogenous anabolic stimulus and did not suppress fibrosis. CLINICAL RELEVANCE This study examines a signaling pathway-based approach to treating poor bone healing in a model of NF1 pseudarthrosis.
Collapse
Affiliation(s)
- J El-Hoss
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - T Cheng
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - E C Carpenter
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - K Sullivan
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - N Deo
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - K Mikulec
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - D G Little
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| | - A Schindeler
- Orthopaedic Research & Biotechnology Unit, The Children's Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145, Australia. E-mail address for J. El-Hoss: . E-mail address for T. Cheng: . E-mail address for E.C. Carpenter: . E-mail address for K. Sullivan: . E-mail address for N. Deo: . E-mail address for K. Mikulec: . E-mail address for D.G. Little: . E-mail address for A. Schindeler:
| |
Collapse
|
76
|
de la Croix Ndong J, Makowski AJ, Uppuganti S, Vignaux G, Ono K, Perrien DS, Joubert S, Baglio SR, Granchi D, Stevenson DA, Rios JJ, Nyman JS, Elefteriou F. Asfotase-α improves bone growth, mineralization and strength in mouse models of neurofibromatosis type-1. Nat Med 2014; 20:904-10. [PMID: 24997609 PMCID: PMC4126855 DOI: 10.1038/nm.3583] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 05/01/2014] [Indexed: 12/20/2022]
Abstract
Mineralization of the skeleton depends on the balance between levels of
pyrophosphate (PPi), an inhibitor of hydroxyapatite formation, and phosphate generated
from PPi breakdown by alkaline phosphatase (ALP). We report here that ablation of
Nf1, encoding the RAS/GTPase–activating protein neurofibromin,
in bone–forming cells leads to supraphysiologic PPi accumulation, caused by a
chronic ERK–dependent increase in genes promoting PPi synthesis and extracellular
transport, namely Enpp1 and Ank. It also prevents
BMP2–induced osteoprogenitor differentiation and, consequently, expression of ALP
and PPi breakdown, further contributing to PPi accumulation. The short stature, impaired
bone mineralization and strength in mice lacking Nf1 in
osteochondroprogenitors or osteoblasts could be corrected by enzyme therapy aimed at
reducing PPi concentration. These results establish neurofibromin as an essential
regulator of bone mineralization, suggest that altered PPi homeostasis contributes to the
skeletal dysplasiae associated with neurofibromatosis type-1 (NF1), and that some of the
NF1 skeletal conditions might be preventable pharmacologically.
Collapse
Affiliation(s)
- Jean de la Croix Ndong
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Alexander J Makowski
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA. [3] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [4] Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Sasidhar Uppuganti
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Guillaume Vignaux
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Koichiro Ono
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [3] Department of Orthopaedics, Nohon Koukan Hospital, Kawasaki, Kanagawa, Japan
| | - Daniel S Perrien
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [3] Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA. [4] Vanderbilt University Institute of Imaging Sciences, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | | | - Serena R Baglio
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - Donatella Granchi
- Laboratory for Orthopedic Pathophysiology and Regenerative Medicine, Istituto Ortopedico Rizzoli, Bologna, Italy
| | - David A Stevenson
- Department of Pediatrics, University of Utah, Salt Lake City, Utah, USA
| | - Jonathan J Rios
- 1] Sarah M. and Charles E. Seay Center for Musculoskeletal Research, Texas Scottish Rite Hospital for Children, Dallas, Texas, USA. [2] Department of Pediatrics, UT Southwestern Medical Center, Dallas, Texas, USA. [3] Eugene McDermott Center for Human Growth &Development, UT Southwestern Medical Center, Dallas, Texas, USA. [4] Department of Orthopaedic Surgery, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jeffry S Nyman
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA. [3] Department of Orthopaedic Surgery &Rehabilitation, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [4] Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, Tennessee, USA
| | - Florent Elefteriou
- 1] Vanderbilt Center for Bone Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [2] Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [3] Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee, USA. [4] Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
77
|
Rhodes SD, Wu X, He Y, Chen S, Yang H, Staser KW, Wang J, Zhang P, Jiang C, Yokota H, Dong R, Peng X, Yang X, Murthy S, Azhar M, Mohammad KS, Xu M, Guise TA, Yang FC. Hyperactive transforming growth factor-β1 signaling potentiates skeletal defects in a neurofibromatosis type 1 mouse model. J Bone Miner Res 2013; 28:2476-89. [PMID: 23703870 PMCID: PMC6774615 DOI: 10.1002/jbmr.1992] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2013] [Revised: 04/29/2013] [Accepted: 05/10/2013] [Indexed: 01/04/2023]
Abstract
Dysregulated transforming growth factor beta (TGF-β) signaling is associated with a spectrum of osseous defects as seen in Loeys-Dietz syndrome, Marfan syndrome, and Camurati-Engelmann disease. Intriguingly, neurofibromatosis type 1 (NF1) patients exhibit many of these characteristic skeletal features, including kyphoscoliosis, osteoporosis, tibial dysplasia, and pseudarthrosis; however, the molecular mechanisms mediating these phenotypes remain unclear. Here, we provide genetic and pharmacologic evidence that hyperactive TGF-β1 signaling pivotally underpins osseous defects in Nf1(flox/-) ;Col2.3Cre mice, a model which closely recapitulates the skeletal abnormalities found in the human disease. Compared to controls, we show that serum TGF-β1 levels are fivefold to sixfold increased both in Nf1(flox/-) ;Col2.3Cre mice and in a cohort of NF1 patients. Nf1-deficient osteoblasts, the principal source of TGF-β1 in bone, overexpress TGF-β1 in a gene dosage-dependent fashion. Moreover, Nf1-deficient osteoblasts and osteoclasts are hyperresponsive to TGF-β1 stimulation, potentiating osteoclast bone resorptive activity while inhibiting osteoblast differentiation. These cellular phenotypes are further accompanied by p21-Ras-dependent hyperactivation of the canonical TGF-β1-Smad pathway. Reexpression of the human, full-length neurofibromin guanosine triphosphatase (GTPase)-activating protein (GAP)-related domain (NF1 GRD) in primary Nf1-deficient osteoblast progenitors, attenuated TGF-β1 expression levels and reduced Smad phosphorylation in response to TGF-β1 stimulation. As an in vivo proof of principle, we demonstrate that administration of the TGF-β receptor 1 (TβRI) kinase inhibitor, SD-208, can rescue bone mass deficits and prevent tibial fracture nonunion in Nf1(flox/-) ;Col2.3Cre mice. In sum, these data demonstrate a pivotal role for hyperactive TGF-β1 signaling in the pathogenesis of NF1-associated osteoporosis and pseudarthrosis, thus implicating the TGF-β signaling pathway as a potential therapeutic target in the treatment of NF1 osseous defects that are refractory to current therapies.
Collapse
Affiliation(s)
- Steven D Rhodes
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, USA; Herman B. Wells Center for Pediatric Research, Indiana University School of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Approaches to treating NF1 tibial pseudarthrosis: consensus from the Children's Tumor Foundation NF1 Bone Abnormalities Consortium. J Pediatr Orthop 2013; 33:269-75. [PMID: 23482262 DOI: 10.1097/bpo.0b013e31828121b8] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Neurofibromatosis 1 (NF1) is an autosomal dominant disorder with various skeletal abnormalities occurring as part of a complex phenotype. Tibial dysplasia, which typically presents as anterolateral bowing of the leg with subsequent fracture and nonunion (pseudarthrosis), is a serious but infrequent osseous manifestation of NF1. Over the past several years, results from clinical and experimental studies have advanced our knowledge of the role of NF1 in bone. On the basis of current knowledge, we propose a number of concepts to consider as a theoretical approach to the optimal management of tibial pseudarthrosis. METHODS A literature review for both clinical treatment and preclinical models for tibial dysplasia in NF1 was performed. Concepts were discussed and developed by experts who participated in the Children's Tumor Foundation sponsored International Bone Abnormalities Consortium meeting in 2011. RESULTS Concepts for a theoretical approach to treating tibial pseudarthrosis include: bone fixation appropriate to achieve stability in any given case; debridement of the "fibrous pseudarthrosis tissue" between the bone segments associated with the pseudarthrosis; creating a healthy vascular bed for bone repair; promoting osteogenesis; controlling overactive bone resorption (catabolism); prevention of recurrence of the "fibrous pseudarthrosis tissue"; and achievement of long-term bone health to prevent recurrence. CONCLUSIONS Clinical trials are needed to assess effectiveness of the wide variation of surgical and pharmacologic approaches currently in practice for the treatment of tibial pseudarthrosis in NF1. LEVEL OF EVIDENCE Level V, expert opinion.
Collapse
|
79
|
Toledo SPA, Lourenço DM, Toledo RA. A differential diagnosis of inherited endocrine tumors and their tumor counterparts. Clinics (Sao Paulo) 2013; 68:1039-56. [PMID: 23917672 PMCID: PMC3715026 DOI: 10.6061/clinics/2013(07)24] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 03/26/2013] [Indexed: 12/15/2022] Open
Abstract
Inherited endocrine tumors have been increasingly recognized in clinical practice, although some difficulties still exist in differentiating these conditions from their sporadic endocrine tumor counterparts. Here, we list the 12 main topics that could add helpful information and clues for performing an early differential diagnosis to distinguish between these conditions. The early diagnosis of patients with inherited endocrine tumors may be performed either clinically or by mutation analysis in at-risk individuals. Early detection usually has a large impact in tumor management, allowing preventive clinical or surgical therapy in most cases. Advice for the clinical and surgical management of inherited endocrine tumors is also discussed. In addition, recent clinical and genetic advances for 17 different forms of inherited endocrine tumors are briefly reviewed.
Collapse
Affiliation(s)
- Sergio P A Toledo
- Division of Endocrinology, Endocrine Genetics Unit (LIM-25), Faculdade de Medicina da Universidade de São Paulo, São Paulo/SP, Brazil.
| | | | | |
Collapse
|
80
|
Greenblatt MB, Shim JH, Glimcher LH. Mitogen-activated protein kinase pathways in osteoblasts. Annu Rev Cell Dev Biol 2013; 29:63-79. [PMID: 23725048 DOI: 10.1146/annurev-cellbio-101512-122347] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Mitogen-activated protein kinases (MAPKs) are ancient signal transducers well characterized as mediators of inflammation and neoplastic transformation. Recent work has expanded our understanding of their developmental functions, particularly in the regulation of bone mass via control of osteoblast differentiation. Here, we review the functions of MAPK pathways in osteoblasts, including a consideration of MAPK substrates. In particular, MAPKs function to regulate the key transcriptional mediators of osteoblast differentiation, with ERK and p38 MAPKs phosphorylating RUNX2, the master regulator of osteoblast differentiation. ERK also activates RSK2, which in turn phosphorylates ATF4, a transcriptional regulator of late-stage osteoblast synthetic functions. The MAP3Ks and MAP2Ks upstream of MAPKs have also been investigated, and significant differences have been found in the wiring of MAPK pathways in osteoblasts relative to other tissues. Thus, the investigation of MAPKs in osteoblasts has both revealed critical mechanisms for the maintenance of bone mass and added to our understanding of how the individual components of MAPK pathways function in concert in a complex in vivo system.
Collapse
Affiliation(s)
- Matthew B Greenblatt
- Department of Pathology, Brigham and Women's Hospital, Boston, Massachusetts 02115;
| | | | | |
Collapse
|
81
|
Giuliani N, Lisignoli G, Magnani M, Racano C, Bolzoni M, Dalla Palma B, Spolzino A, Manferdini C, Abati C, Toscani D, Facchini A, Aversa F. New insights into osteogenic and chondrogenic differentiation of human bone marrow mesenchymal stem cells and their potential clinical applications for bone regeneration in pediatric orthopaedics. Stem Cells Int 2013; 2013:312501. [PMID: 23766767 PMCID: PMC3676919 DOI: 10.1155/2013/312501] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) are pluripotent adult stem cells capable of being differentiated into osteoblasts, adipocytes, and chondrocytes. The osteogenic differentiation of hMSCs is regulated either by systemic hormones or by local growth factors able to induce specific intracellular signal pathways that modify the expression and activity of several transcription factors. Runt-related transcription factor 2 (Runx2) and Wnt signaling-related molecules are the major factors critically involved in the osteogenic differentiation process by hMSCs, and SRY-related high-mobility-group (HMG) box transcription factor 9 (SOX9) is involved in the chondrogenic one. hMSCs have generated a great interest in the field of regenerative medicine, particularly in bone regeneration. In this paper, we focused our attention on the molecular mechanisms involved in osteogenic and chondrogenic differentiation of hMSC, and the potential clinical use of hMSCs in osteoarticular pediatric disease characterized by fracture nonunion and pseudarthrosis.
Collapse
Affiliation(s)
- Nicola Giuliani
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Gina Lisignoli
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale e Laboratorio RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Marina Magnani
- Paediatric Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Costantina Racano
- Paediatric Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Marina Bolzoni
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Benedetta Dalla Palma
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Angelica Spolzino
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Cristina Manferdini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale e Laboratorio RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Caterina Abati
- Paediatric Orthopaedics and Traumatology, Rizzoli Orthopaedic Institute, Via GC Pupilli 1, 40136 Bologna, Italy
| | - Denise Toscani
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| | - Andrea Facchini
- SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale e Laboratorio RAMSES, Rizzoli Orthopaedic Institute, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Franco Aversa
- Hematology, Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, 43126 Parma, Italy
| |
Collapse
|
82
|
Ono K, Karolak MR, Ndong JDLC, Wang W, Yang X, Elefteriou F. The ras-GTPase activity of neurofibromin restrains ERK-dependent FGFR signaling during endochondral bone formation. Hum Mol Genet 2013; 22:3048-62. [PMID: 23571107 DOI: 10.1093/hmg/ddt162] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The severe defects in growth plate development caused by chondrocyte extracellular signal-regulated protein kinases 1 and 2 (ERK1/2) gain or loss-of-function suggest that tight spatial and temporal regulation of mitogen-activated protein kinase signaling is necessary to achieve harmonious growth plate elongation and structure. We provide here evidence that neurofibromin, via its Ras guanosine triphosphatase -activating activity, controls ERK1/2-dependent fibroblast growth factor receptor (FGFR) signaling in chondrocytes. We show first that neurofibromin is expressed in FGFR-positive prehypertrophic and hypertrophic chondrocytes during growth plate endochondral ossification. Using mice lacking neurofibromin 1 (Nf1) in type II collagen-expressing cells, (Nf1col2(-/-) mutant mice), we then show that lack of neurofibromin in post-mitotic chondrocytes triggers a number of phenotypes reminiscent of the ones observed in mice characterized by FGFR gain-of-function mutations. Those include dwarfism, constitutive ERK1/2 activation, strongly reduced Ihh expression and decreased chondrocyte proliferation and maturation, increased chondrocytic expression of Rankl, matrix metalloproteinase 9 (Mmp9) and Mmp13 and enhanced growth plate osteoclastogenesis, as well as increased sensitivity to caspase-9 mediated apoptosis. Using wildtype (WT) and Nf1(-/-) chondrocyte cultures in vitro, we show that FGF2 pulse-stimulation triggers rapid ERK1/2 phosphorylation in both genotypes, but that return to the basal level is delayed in Nf1(-/-) chondrocytes. Importantly, in vivo ERK1/2 inhibition by daily injection of a recombinant form of C-type natriuretic peptide to post-natal pups for 18 days was able to correct the short stature of Nf1col2(-/-) mice. Together, these results underscore the requirement of neurofibromin and ERK1/2 for normal endochondral bone formation and support the notion that neurofibromin, by restraining RAS-ERK1/2 signaling, is a negative regulator of FGFR signaling in differentiating chondrocytes.
Collapse
Affiliation(s)
- Koichiro Ono
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232, USA
| | | | | | | | | | | |
Collapse
|
83
|
Arrington DK, Danehy AR, Peleggi A, Proctor MR, Irons MB, Ullrich NJ. Calvarial defects and skeletal dysplasia in patients with neurofibromatosis Type 1. J Neurosurg Pediatr 2013; 11:410-6. [PMID: 23414129 DOI: 10.3171/2013.1.peds12409] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
OBJECT Skull defects, including sphenoid dysplasia and calvarial defects, are rare but distinct findings in patients with neurofibromatosis Type 1 (NF1). The underlying pathophysiology is unclear. The goal of this study was to identify the clinical characteristics and natural history of skull defects in patients with NF1. METHODS An electronic search engine of medical records was used to identify patients with NF1 and bony skull anomalies. All clinical, radiographic, pathology, and operative reports were reviewed. The relationship between bony anomalies and significant clinical associations was evaluated. This study received institutional review board approval. RESULTS Twenty-one patients were identified. The mean age at NF1 diagnosis was 4.2 years. The mean age at skull defect diagnosis was 8.8 years (9.7 years in the sphenoid wing dysplasia group and 11.9 years in the calvarial defect group). Sphenoid dysplasia was associated with a plexiform neurofibroma or dural ectasia in 73.3% and 80.0% of cases, respectively. Calvarial defects were associated with a plexiform neurofibroma or dural ectasia in 66.7% and 33.3% of patients, respectively. An absence of either an associated neurofibroma or ectasia was not noted in any patient with sphenoid wing dysplasia or 25.0% of those with calvarial defects. In 6 patients, both types of skull defects presented simultaneously. Serial imaging studies were obtained for a mean follow-up time of 7.5 years (range 0.4-20.0 years). Of these patients with serial imaging, radiographic progression was found in 60% of cases of calvarial defects and 56% of cases of sphenoid wing dysplasia. Two patients underwent surgical repair of a skull defect, and both required repeat procedures. CONCLUSIONS The majority of skull defects in patients with NF1 were associated with an adjacent structural lesion, such as a plexiform neurofibroma or dural ectasia. This findings from this cohort also support the concept of progression in defect size in more than half of the patients. Potential mechanisms by which these secondary lesions contribute to pathogenesis of the bony defect may include changes in the bony microenvironment. A better understanding of the pathophysiology of skull defects will help guide detection, improve treatment and outcome, and may contribute to the understanding of the pathogenesis of bony lesions in NF1.
Collapse
Affiliation(s)
- Daniel K Arrington
- Departments of Neurology, Boston Children's Hospital, 300 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | | | | | |
Collapse
|
84
|
Gutmann DH, Blakeley JO, Korf BR, Packer RJ. Optimizing biologically targeted clinical trials for neurofibromatosis. Expert Opin Investig Drugs 2013; 22:443-62. [PMID: 23425047 PMCID: PMC4009992 DOI: 10.1517/13543784.2013.772979] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
INTRODUCTION The neurofibromatoses (neurofibromatosis type 1, NF1 and neurofibromatosis type 2, NF2) comprise the most common inherited conditions in which affected children and adults develop tumors of the central and peripheral nervous system. In this review, the authors discuss how the establishment of the Neurofibromatosis Clinical Trials Consortium (NFCTC) has positively impacted on the design and execution of treatment studies for individuals with NF1 and NF2. AREAS COVERED Using an extensive PUBMED search in collaboration with select NFCTC members expert in distinct NF topics, the authors discuss the clinical features of NF1 and NF2, the molecular biology of the NF1 and NF2 genes, the development and application of clinically relevant Nf1 and Nf2 genetically engineered mouse models and the formation of the NFCTC to enable efficient clinical trial design and execution. EXPERT OPINION The NFCTC has resulted in a more seamless integration of mouse preclinical and human clinical trials efforts. Leveraging emerging enabling resources, current research is focused on identifying subtypes of tumors in NF1 and NF2 to deliver the most active compounds to the patients most likely to respond to the targeted therapy.
Collapse
Affiliation(s)
- David H Gutmann
- Washington University School of Medicine, Department of Neurology and Washington University Neurofibromatosis Center, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | | | | | |
Collapse
|
85
|
George-Abraham JK, Martin LJ, Kalkwarf HJ, Rieley MB, Stevenson DA, Viskochil DH, Hopkin RJ, Stevens AM, Hanson H, Schorry EK. Fractures in children with neurofibromatosis type 1 from two NF clinics. Am J Med Genet A 2013; 161A:921-6. [PMID: 23529831 DOI: 10.1002/ajmg.a.35541] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2012] [Accepted: 06/01/2012] [Indexed: 01/01/2023]
Abstract
Neurofibromatosis type 1 (NF1) is an autosomal dominant genetic disorder with osseous abnormalities occurring in up to one-third of patients. Several studies have documented osteopenia in both children and adults with NF1; however, the significance of lower bone mineral density (BMD) in relationship to fracture incidence is not well elucidated in NF1, particularly in children. We undertook a retrospective study to determine prevalence and location of fractures in children and adolescents with NF1, ages 5-20 years, using a standardized questionnaire. We surveyed 256 individuals with NF1 from two multidisciplinary NF centers and 178 controls without NF1 of similar ages and sex. Participants with known long bone dysplasia (LBD) were analyzed separately. Data collected included numbers and location of fractures, dietary calcium intake, and physical activity levels. There was no difference in prevalence of ever having a fracture between the NF1 group without LBD (22%) and the control group (25%); median number of fractures also did not differ. There were significant differences in fracture location with a higher frequency of fractures of the lower extremities in NF1 individuals without LBD compared to controls. Both NF1 cohorts had lower rates of physical activity than controls (P < 0.0001). Our data demonstrate that the likelihood of having had a fracture is not higher in young NF1 individuals without LBD in comparison to healthy controls. The lower physical activity level may have a "protective effect" for those with NF1, thus keeping their fracture incidence lower than expected for their relative degree of osteopenia.
Collapse
Affiliation(s)
- Jaya K George-Abraham
- Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Soucy EA, van Oppen D, Nejedly NL, Gao F, Gutmann DH, Hollander AS. Height assessments in children with neurofibromatosis type 1. J Child Neurol 2013; 28:303-7. [PMID: 22752476 PMCID: PMC3947790 DOI: 10.1177/0883073812446310] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Previous studies have suggested that children with neurofibromatosis type 1 (NF1) are shorter than their unaffected counterparts. Unfortunately, these reports did not consider other contributing factors that might also influence short stature. The purpose of the current study was to characterize the genetic influence of NF1 on the growth of children. Height data were measured and recorded for 170 patients, whereas parental measurements were obtained for 61 patients to calculate sex-corrected mid-parental target heights. Children with NF1 had population mean height and mid-parental height z scores statistically different from the general population. Importantly, these differences were pronounced when neither parent had NF1 but were not significant when one of the parents had NF1. Moreover, height z scores for children with NF1 were also statistically different than their unaffected siblings. Collectively, these data establish a clear effect of a germline NF1 gene mutation on stature in children with NF1.
Collapse
Affiliation(s)
- Elizabeth A. Soucy
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Dorothy van Oppen
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Nicole L. Nejedly
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| | - Feng Gao
- Division of Biostatistics, Washington University School of Medicine, St. Louis, MO 63110
| | - David H. Gutmann
- Department of Neurology, Washington University School of Medicine, St. Louis, MO 63110
| | - Abby S. Hollander
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110
| |
Collapse
|
87
|
Abstract
The likelihood of suffering a bone fracture is not solely predicated on areal bone mineral density. As people age, there are numerous changes to the skeleton occurring at multiple length scales (from millimeters to submicron scales) that reduce the ability of bone to resist fracture. Herein is a review of the current knowledge about the role of the extracellular matrix (ECM) in this resistance, with emphasis on engineering principles that characterize fracture resistance beyond bone strength to include bone toughness and fracture toughness. These measurements of the capacity to dissipate energy and to resist crack propagation during failure precipitously decline with age. An age-related loss in collagen integrity is strongly associated with decreases in these mechanical properties. One potential cause for this deleterious change in the ECM is an increase in advanced glycation end products, which accumulate with aging through nonenzymatic collagen crosslinking. Potential regulators and diagnostic tools of the ECM with respect to fracture resistance are also discussed.
Collapse
Affiliation(s)
- Jeffry S Nyman
- Department of Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 27212, USA.
| | | |
Collapse
|
88
|
Doyle JJ, Gerber EE, Dietz HC. Matrix-dependent perturbation of TGFβ signaling and disease. FEBS Lett 2012; 586:2003-15. [PMID: 22641039 PMCID: PMC3426037 DOI: 10.1016/j.febslet.2012.05.027] [Citation(s) in RCA: 105] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2012] [Revised: 05/13/2012] [Accepted: 05/15/2012] [Indexed: 02/07/2023]
Abstract
Transforming growth factor beta (TGFβ) is a multipotent cytokine that is sequestered in the extracellular matrix (ECM) through interactions with a number of ECM proteins. The ECM serves to concentrate latent TGFβ at sites of intended function, to influence the bioavailability and/or function of TGFβ activators, and perhaps to regulate the intrinsic performance of cell surface effectors of TGFβ signal propagation. The downstream consequences of TGFβ signaling cascades in turn provide feedback modulation of the ECM. This review covers recent examples of how genetic mutations in constituents of the ECM or TGFβ signaling cascade result in altered ECM homeostasis, cellular performance and ultimately disease, with an emphasis on emerging therapeutic strategies that seek to capitalize on this refined mechanistic understanding.
Collapse
|
89
|
Alanne MH, Siljamäki E, Peltonen S, Väänänen K, Windle JJ, Parada LF, Määttä JA, Peltonen J. Phenotypic characterization of transgenic mice harboring Nf1+/− or Nf1−/− osteoclasts in otherwise Nf1+/+ background. J Cell Biochem 2012; 113:2136-46. [DOI: 10.1002/jcb.24088] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
90
|
Elefteriou F, Yang X. Genetic mouse models for bone studies--strengths and limitations. Bone 2011; 49:1242-54. [PMID: 21907838 PMCID: PMC3331798 DOI: 10.1016/j.bone.2011.08.021] [Citation(s) in RCA: 99] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Revised: 08/15/2011] [Accepted: 08/18/2011] [Indexed: 11/25/2022]
Abstract
Mice have become a preferred model system for bone research because of their genetic and pathophysiological similarities to humans: a relatively short reproductive period, leading to relatively low cost of maintenance and the availability of the entire mouse genome sequence information. The success in producing the first transgenic mouse line that expressed rabbit β-globin protein in mouse erythrocytes three decades ago marked the beginning of the use of genetically engineered mice as model system to study human diseases. Soon afterward the development of cultured pluripotent embryonic stem cells provided the possibility of gene replacement or gene deletion in mice. These technologies have been critical to identify new genes involved in bone development, growth, remodeling, repair, and diseases, but like many other approaches, they have limitations. This review will introduce the approaches that allow the generation of transgenic mice and global or conditional (tissue-specific and inducible) mutant mice. A list of the various promoters used to achieve bone-specific gene deletion or overexpression is included. The limitations of these approaches are discussed, and general guidelines related to the analysis of genetic mouse models are provided.
Collapse
Affiliation(s)
- Florent Elefteriou
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| | - Xiangli Yang
- Vanderbilt University Medical Center, Department of Medicine, Vanderbilt Center for Bone Biology, 1235H Light Hall, Nashville, TN 37232-0575, USA
| |
Collapse
|