1
|
Meyfarth SRS, Antunes LAA, da Silva Tavares J, Guimarães LDS, da Silva EAB, Baratto-Filho F, Küchler EC, Silva-Sousa AC, Sousa-Neto MD, Antunes LS. Single nucleotide polymorphisms in inducible nitric oxide synthase gene are not associated with persistent apical periodontitis. AUST ENDOD J 2023; 49:648-656. [PMID: 37724624 DOI: 10.1111/aej.12796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Accepted: 09/04/2023] [Indexed: 09/21/2023]
Abstract
The aim of this study was to investigate whether there is an association between inducible in single nucleotide polymorphisms in nitric oxide synthase (rs2297518 and rs2779249) and persistent apical periodontitis. A total of 291 Brazilian subjects were included: 125 with signs/symptoms of persistent apical periodontitis and 166 with root canal-treated teeth exhibiting healthy perirradicular tissues. Endodontically treated patients were followed up after 1 year. The two single nucleotide polymorphisms in nitric oxide synthase were analysed using real-time polymerase chain reaction. Chi-square test and odds ratio with 95% confidence intervals were performed to compare genotype distributions between 'healed' and 'persistent apical periodontitis' groups (p < 0.05). Logistic regression analysis was used to evaluate SNP-SNP interactions. The allele and genotype distributions for the polymorphisms between the persistent apical periodontitis and healed groups were not statistically significant (p > 0.05). In the logistic regression analysis, the polymorphisms were not associated with persistent apical periodontitis and SNP-SNP interactions.
Collapse
Affiliation(s)
| | - Lívia Azeredo Alves Antunes
- Postgraduate Program, School of Dentistry, Fluminense Federal University, Niterói, Brazil
- Clinical Research Unit, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program, School of Dentistry, Fluminense Federal University, Nova Friburgo, Brazil
- Specific Formation Department, School of Dentistry of Nova Friburgo, Fluminense Federal University, Nova Friburgo, Brazil
| | | | | | | | | | | | - Alice Corrêa Silva-Sousa
- Restorative Dentistry Department, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Manoel Damião Sousa-Neto
- Restorative Dentistry Department, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, Brazil
| | - Leonardo Santos Antunes
- Postgraduate Program, School of Dentistry, Fluminense Federal University, Niterói, Brazil
- Clinical Research Unit, Fluminense Federal University, Niterói, Brazil
- Postgraduate Program, School of Dentistry, Fluminense Federal University, Nova Friburgo, Brazil
- Specific Formation Department, School of Dentistry of Nova Friburgo, Fluminense Federal University, Nova Friburgo, Brazil
| |
Collapse
|
2
|
Petean IBF, Silva-Sousa AC, Cronenbold TJ, Mazzi-Chaves JF, Silva LABD, Segato RAB, Castro GAPD, Kuchler EC, Paula-Silva FWG, Damião Sousa-Neto M. Genetic, Cellular and Molecular Aspects involved in Apical Periodontitis. Braz Dent J 2022; 33:1-11. [PMID: 36043561 PMCID: PMC9645190 DOI: 10.1590/0103-6440202205113] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 08/03/2022] [Indexed: 11/26/2022] Open
Abstract
The development, establishment and repair of apical periodontitis (AP) is
dependent of several factors, which include host susceptibility, microbial
infection, immune response, quality of root canal treatment and organism's
ability to repair. The understanding of genetic contributions to the risk of
developing AP and presenting persistent AP has been extensively explored in
modern Endodontics. Thus, this article aims to provide a review of the
literature regarding the biochemical mediators involved in immune response
signaling, osteoclastogenesis and bone neoformation, as the genetic components
involved in the development and repair of AP. A narrative review of the
literature was performed through a PUBMED/MEDLINE search and a hand search of
the major AP textbooks. The knowledge regarding the cells, receptors and
molecules involved in the host's immune-inflammatory response during the
progression of AP added to the knowledge of bone biology allows the
identification of factors inherent to the host that can interfere both in the
progression and in the repair of these lesions. The main outcomes of studies
evaluated in the review that investigated the correlation between genetic
polymorphisms and AP in the last five years, demonstrate that genetic factors of
the individual are involved in the success of root canal treatment. The
discussion of this review gives subsides that may help to glimpse the
development of new therapies based on the identification of therapeutic targets
and the development of materials and techniques aimed at acting at the molecular
level for clinical, radiographic and histological success of root canal
treatment.
Collapse
Affiliation(s)
- Igor Bassi Ferreira Petean
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Brazil
| | - Alice Corrêa Silva-Sousa
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Brazil
| | | | | | - Lea Assed Bezerra da Silva
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Raquel Assed Bezerra Segato
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | | | - Erika Calvano Kuchler
- Department of Pediatric Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Department of Orthodontics, University of Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany
| | | | - Manoel Damião Sousa-Neto
- Department of Restorative Dentistry, School of Dentistry of Ribeirão Preto, University of São Paulo, Brazil
| |
Collapse
|
3
|
Aglan HA, Fouad-Elhady EA, Hassan RE, Sabry GM, Ahmed HH. Nanoplatforms for Promoting Osteogenesis in Ovariectomy-Induced
Osteoporosis in the Experimental Model. CURRENT NANOMEDICINE 2022; 12:44-62. [DOI: 10.2174/2468187312666220217104650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2021] [Revised: 12/19/2021] [Accepted: 01/12/2022] [Indexed: 01/05/2025]
Abstract
Background:
Osteoporosis is a debilitating bone ailment characterized by the obvious loss of bone mass and bone microarchitecture impairment.
Objective:
This study aimed to illuminate the in vivo usefulness of nanotechnology as a treatment for osteoporosis via analyzing the effectiveness of nano-hydroxyapatite (nHa), nano-hydroxy- apatite/chitosan (nHa/C), and nano-hydroxyapatite/silver (nHa/S) in mitigation of osteoporosis in ovariectomized rats.
Method:
The characterization of the nHa, nHa/C, and nHa/S was carried out using TEM, SEM, FTIR, and Zeta potential measurements. This in vivo study included 48 adult female rats that were randomized into six groups (8 rats/group): (1) Sham-operated control, (2) osteoporotic, (3) nHa, (4) nHa/C, (5) nHa/S, and (6) Fosamax®. Serum osterix level was quantified using ELISA. Femur bone morphogenetic protein 2 and SMAD1 mRNA levels were evaluated by qPCR. The femur bones were scanned by DEXA for measurement of bone mineral density and bone mineral content. In ad-dition, a histopathological examination of femur bones was performed.
Results:
The present approach denoted that the treatment with nHa, nHa/C, or nHa/S yields a signif-icant rise in serum level of osterix and mRNA levels of bone morphogenetic protein 2 and SMAD1 as well as significant enhancements of bone tissue minerals.
Conclusion:
The findings affirmed the potency of nHa, nHa/C, and nHa/S as auspicious nanoplat-forms for repairing bone defects in the osteoporotic rat model. The positive effect of the inspected nanoformulations arose from bone formation indicators in serum and tissue, and additionally, the reinforcement of bone density and content, which were verified by the histopathological description of bone tissue sections.
Collapse
Affiliation(s)
- Hadeer A. Aglan
- Hormones Department, Medicine and Clinical Studies Research Institute, National Research Centre, Giza, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| | | | - Rasha E. Hassan
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Gilane M. Sabry
- Biochemistry Department, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Hanaa H. Ahmed
- Hormones Department, Medicine and Clinical Studies Research Institute, National Research Centre, Giza, Egypt
- Stem Cells Lab, Center of Excellence for Advanced Sciences, National Research Centre, Giza, Egypt
| |
Collapse
|
4
|
Yang H, Liang Y, Cao Y, Cao Y, Fan Z. Homeobox C8 inhibited the osteo-/dentinogenic differentiation and migration ability of stem cells of the apical papilla via activating KDM1A. J Cell Physiol 2020; 235:8432-8445. [PMID: 32246725 DOI: 10.1002/jcp.29687] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Revised: 03/18/2020] [Accepted: 03/23/2020] [Indexed: 12/13/2022]
Abstract
Enhancing the functions of mesenchymal stem cells (MSCs) is considered a potential approach for promoting tissue regeneration. In the present study, we investigate the role of HOXC8 in regulating differentiation and migration by using stem cells of the apical papilla (SCAPs). Our results showed that overexpression of HOXC8 suppressed the osteo-/dentinogenic differentiation, as detected by measuring alkaline phosphatase activity, in vitro mineralization, and the expressions of dentin sialophosphoprotein, dentin matrix acidic phosphoprotein 1, bone sialoprotein, runt-related transcription factor 2, and osterix in SCAPs, and inhibited in vivo osteo-/dentinogenesis of SCAPs. In addition, knockdown of HOXC8 promoted the osteo-/dentinogenic differentiation potentials of SCAPs. Mechanically, HOXC8 enhanced KDM1A transcription by directly binding to its promoter. HOXC8 and KDM1A also inhibited the migration and chemotaxis abilities of SCAPs. To sum up, HOXC8 negatively regulated the osteo-/dentinogenic differentiation and migration abilities of SCAPs by directly enhancing KDM1A transcription and indicated that HOXC8 and KDM1A could serve as potential targets for enhancing dental MSC mediated tissue regeneration.
Collapse
Affiliation(s)
- Haoqing Yang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Stomatology Hospital, Capital Medical University, Beijing, China
| | - Yuncun Liang
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Stomatology Hospital, Capital Medical University, Beijing, China
| | - Yangyang Cao
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Stomatology Hospital, Capital Medical University, Beijing, China
| | - Yu Cao
- Department of General Dentistry, School of Stomatology, Beijing Stomatology Hospital, Capital Medical University, Beijing, China
| | - Zhipeng Fan
- Beijing Key Laboratory of Tooth Regeneration and Function Reconstruction, School of Stomatology, Beijing Stomatology Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
5
|
Li Q, Han G, Liu D, Zhou Y. Force-induced decline of TEA domain family member 1 contributes to osteoclastogenesis via regulation of Osteoprotegerin. Arch Oral Biol 2019; 100:23-32. [PMID: 30771694 DOI: 10.1016/j.archoralbio.2019.01.020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 01/28/2019] [Accepted: 01/30/2019] [Indexed: 01/06/2023]
Abstract
OBJECTIVE This study aims to investigate the responsiveness of transcription factor TEA domain family member 1 (TEAD1) to mechanical force and its impact on osteoclastogenesis as well as expression of Osteoprotegerin (OPG), an inhibitor for osteoclastogenesis playing crucial roles in mechanical stress-induced bone remodeling and orthodontic tooth movement (OTM). METHODS We first analyzed the correlation between several transcription factors and OPG expression in human periodontal ligament cells (PDLCs). Then dynamic expression changes of TEAD1 with force application were analyzed due to its high correlation with OPG. Loss-of-function experiments were performed to demonstrate the role of TEAD1 in regulation of RANKL/OPG, as well as osteoclastogenesis by tartrate-resistant acid phosphatase (TRAP) staining. Combination of bioinformatics analyzes and chromatin immunoprecipitation assay was utilized to investigate occupancy of TEAD1 on the enhancer elements of OPG and the dynamic change in response to force stimuli. Involvement of Hippo signaling in regulation of OPG was further demonstrated by pharmacologic inhibitors of several components. RESULTS Expression of TEAD1 highly correlates with that of OPG and decreases in response to mechanical force in human PDLCs. Knockdown of TEAD1 downregulates expression of OPG and promotes osteoclast differentiation. Mechanical force induced decreased binding of TEAD1 on an enhancer element ˜22 kilobases upstream of OPG promoter. OPG was also affected by pharmaceutical disruption of Hippo signaling pathway. CONCLUSIONS TEAD1 is a novel mechano-responsive gene and plays an important role in force-induced osteoclastogenesis, which is dependent, as least partially, on transcriptional regulation of OPG.
Collapse
Affiliation(s)
- Qian Li
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| | - Gaofeng Han
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Dawei Liu
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China
| | - Yanheng Zhou
- Department of Orthodontics, Peking University School and Hospital of Stomatology, National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing Key Laboratory of Digital Stomatology, Beijing, China.
| |
Collapse
|
6
|
MacFarlane EG, Haupt J, Dietz HC, Shore EM. TGF-β Family Signaling in Connective Tissue and Skeletal Diseases. Cold Spring Harb Perspect Biol 2017; 9:cshperspect.a022269. [PMID: 28246187 DOI: 10.1101/cshperspect.a022269] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The transforming growth factor β (TGF-β) family of signaling molecules, which includes TGF-βs, activins, inhibins, and numerous bone morphogenetic proteins (BMPs) and growth and differentiation factors (GDFs), has important functions in all cells and tissues, including soft connective tissues and the skeleton. Specific TGF-β family members play different roles in these tissues, and their activities are often balanced with those of other TGF-β family members and by interactions with other signaling pathways. Perturbations in TGF-β family pathways are associated with numerous human diseases with prominent involvement of the skeletal and cardiovascular systems. This review focuses on the role of this family of signaling molecules in the pathologies of connective tissues that manifest in rare genetic syndromes (e.g., syndromic presentations of thoracic aortic aneurysm), as well as in more common disorders (e.g., osteoarthritis and osteoporosis). Many of these diseases are caused by or result in pathological alterations of the complex relationship between the TGF-β family of signaling mediators and the extracellular matrix in connective tissues.
Collapse
Affiliation(s)
- Elena Gallo MacFarlane
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205
| | - Julia Haupt
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205.,Howard Hughes Medical Institute, Bethesda, Maryland 21205
| | - Eileen M Shore
- Department of Orthopedic Surgery, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Center for Research in FOP and Related Disorders, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104.,Department of Genetics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania 19104
| |
Collapse
|
7
|
Choucair N, Rajab M, Mégarbané A, Chouery E. Homozygous microdeletion of the ERI1 and MFHAS1 genes in a patient with intellectual disability, limb abnormalities, and cardiac malformation. Am J Med Genet A 2017; 173:1955-1960. [PMID: 28488351 DOI: 10.1002/ajmg.a.38271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 04/03/2017] [Indexed: 02/03/2023]
Abstract
A male child, born from consanguineous parents and having intellectual disability, short stature, dysmorphic facial features, synpolydactyly, and cardiac malformations is reported. Chromosomal microarray analysis showed that the patient presents with an 8p23.1 homozygous deletion, containing the microRNA miR-4660, the exoribonuclease 1 (ERI1), and malignant fibrous histiocytoma amplified sequence 1 (MFHAS1) genes. The microRNA miR-4660 has no known function. MFHAS1 is an immunomodulatory protein involved in Toll-like receptor signaling, erythropoiesis, and cancer. ERI1 is a ribonuclease involved in RNA metabolism and is required for the correct patterning of the skeleton by defining the HOXC8 expression. We discuss the involvement of these deleted genes to the patient's features and highlight differential diagnoses with syndromes implicating limb extremity abnormalities such as synpolydactyly, including the monosomy 8p.
Collapse
Affiliation(s)
- Nancy Choucair
- Unité de Génétique Médicale, Faculté de Médecine, Université Saint-Joseph, Beirut, Lebanon
| | - Mariam Rajab
- Department of Pediatrics, Makassed General Hospital, Beirut, Lebanon
| | | | - Eliane Chouery
- Unité de Génétique Médicale, Faculté de Médecine, Université Saint-Joseph, Beirut, Lebanon
| |
Collapse
|
8
|
Huang HM, Li XL, Tu SQ, Chen XF, Lu CC, Jiang LH. Effects of Roughly Focused Extracorporeal Shock Waves Therapy on the Expressions of Bone Morphogenetic Protein-2 and Osteoprotegerin in Osteoporotic Fracture in Rats. Chin Med J (Engl) 2017; 129:2567-2575. [PMID: 27779163 PMCID: PMC5125335 DOI: 10.4103/0366-6999.192776] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
BACKGROUND Roughly focused extracorporeal shock waves therapy (ESWT) is characterized by a wide focal area, a large therapy zone, easy positioning, and less pain during treatment. The purpose of this study was to investigate the effects of roughly focused ESWT on the expression of osteoprotegerin (OPG) and bone morphogenetic protein-2 (BMP-2) in osteoporotic fractures in rats. METHODS Seventy-two female Sprague-Dawley (SD) rats, 3 months old, were divided into sham-operated group (n = 6) and an ovariectomized (OVX) group (n = 66). Sixty OVX SD rats were used as a model of double proximal tibial osteotomy and inner fixation. The osteotomy site in the left tibia was treated with roughly focused ESWT once at an energy density of 0.26 mJ/mm2, 60 doses/min, and 2000 pact quantities. The contralateral right tibia was left untreated and served as a control. Expression of OPG and BMP-2 in the callus of the osteoporotic fracture area was assessed using immunohistochemistry, real-time polymerase chain reaction (PCR), and Western blotting analysis. RESULTS Bone mineral density (BMD) at the proximal tibia, femur, and L5 spine was significantly reduced after ovariectomy. BMD of proximal tibia was 12.9% less in the OVX group than that in the sham-operated group. Meanwhile, bilateral oophorectomy resulted in a lower trabecular bone volume fraction (BV/TV) in the proximal tibia of the sham-OVX animals. Three months after bilateral oophorectomy, BV/TV was 14.29% of baseline BV/TV in OVX legs versus 45.91% in the sham-OVX legs (P < 0.001). These data showed that the SD rats became a suitable model of osteoporosis, 3 months after they were OVX. Immunohistochemical analysis showed higher levels of BMP-2 and OPG expression in the treatment group than those in the control group. Compared with the contralateral controls, decreased expression of OPG and BMP-2 at 3 days after roughly focused ESWT, followed by a later increase at 7 days, was indicated by real-time PCR and Western blotting analysis. The OPG messenger RNA (mRNA) expression levels peaked at 6 weeks after the shock wave treatment, paired with a much earlier (at 4 weeks) increase of BMP-2, and declined close to normal at 8 weeks. CONCLUSIONS Roughly focused ESWT may promote the expression of OPG and BMP-2 in the osteoporotic fracture area in rats. BMP-2 and OPG may act synergistically and may lead to a significant enhancement of bone formation and remodeling.
Collapse
Affiliation(s)
- Hai-Ming Huang
- Department of Orthopedic Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Xiao-Lin Li
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Shu-Qiang Tu
- Department of Orthopedic Surgery, Nanchang Hongdu Hospital of Traditional Chinese Medicine, Nanchang, Jiangxi 330006, China
| | - Xiao-Feng Chen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Chang-Chun Lu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University, Shanghai 200233, China
| | - Liang-Hua Jiang
- Department of Orthopedic Surgery, Kunshan First People's Hospital, Suzhou, Jiangsu 215300, China
| |
Collapse
|
9
|
Soliman S, Ahmed M. The Effect of Orthognathic Surgery on Osteoprotegerin as Immunological Caliper of Bone Healing. Open Access Maced J Med Sci 2016; 4:705-708. [PMID: 28028419 PMCID: PMC5175527 DOI: 10.3889/oamjms.2016.123] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 10/29/2016] [Accepted: 10/30/2016] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND: Osteoprotegerin (OPG) is considered to be the cytokine that plays an important role in the healing process. OPG regulates bone cell biology, osteoblast–osteoclast, bone-immune cross-talk and maintenance of bone mass. It plays an important role in the development, induction, and repair of bone. Orthognathic surgery as multiples segmental osteotomies has been taken as a model surgery to assess the changes in osteoprotegerin levels in the post-operative bone healing period. AIM: The aim of the study was to evaluate OPG as immunological caliper of bone healing. MATERIAL AND METHODS: OPG was evaluated in nine patients seeking orthognathic surgery. Patients were examined and checked to be medically and immunologically free prior to surgery. Blood samples were collected immediate pre-operative as control group and for six weeks post-operative as study group. RESULTS: Data were collected from nine consecutive patients. The results showed higher levels of OPG. it showed significant increase in the immediate post-operative value (p = 0.001) which started to increase gradually during the six weeks (p < 0.001). CONCLUSIONS: Significant higher levels of OPG during the healing period of orthognathic surgery suggest the its use as immunological caliper of bone healing.
Collapse
Affiliation(s)
- Sara Soliman
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Cairo University, Cairo, Egypt
| | - Mamdouh Ahmed
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Cairo University, Cairo, Egypt
| |
Collapse
|
10
|
The biological function of type I receptors of bone morphogenetic protein in bone. Bone Res 2016; 4:16005. [PMID: 27088043 PMCID: PMC4820739 DOI: 10.1038/boneres.2016.5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 02/04/2016] [Accepted: 02/20/2016] [Indexed: 12/13/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) have multiple roles in skeletal development, homeostasis and regeneration. BMPs signal via type I and type II serine/threonine kinase receptors (BMPRI and BMPRII). In recent decades, genetic studies in humans and mice have demonstrated that perturbations in BMP signaling via BMPRI resulted in various diseases in bone, cartilage, and muscles. In this review, we focus on all three types of BMPRI, which consist of activin-like kinase 2 (ALK2, also called type IA activin receptor), activin-like kinase 3 (ALK3, also called BMPRIA), and activin-like kinase 6 (ALK6, also called BMPRIB). The research areas covered include the current progress regarding the roles of these receptors during myogenesis, chondrogenesis, and osteogenesis. Understanding the physiological and pathological functions of these receptors at the cellular and molecular levels will advance drug development and tissue regeneration for treating musculoskeletal diseases and bone defects in the future.
Collapse
|
11
|
Abstract
Osteoprotegerin (OPG) is a key regulator of bone remodeling. Mutations in OPG are involved in a variety of human diseases. We have shown that cochlear spiral ganglion cells secrete OPG at high levels and lack of OPG causes sensorineural hearing loss in addition to the previously described conductive hearing loss. In order to study the regulation of OPG expression, we conducted a database search on regulatory elements in the promoter region of the OPG gene, and identified two potential GATA-3 binding sites. Using luciferase assays and site directed mutagenesis, we demonstrate that these two elements are GATA-3 responsive and support GATA-3 transactivation in human HEK and HeLa cells. The expression of wild type GATA-3 activated OPG mRNA and protein expression, while the expression of a dominant negative mutant of GATA-3 or a GATA-3 shRNA construct reduced OPG mRNA and protein levels. GATA-3 deficient cells generated by expressing a GATA-3 shRNA construct were sensitive to apoptosis induced by etoposide and TNF-α. This apoptotic effect could be partly prevented by the co-treatment with exogenous OPG. Our results suggest new approaches to rescue diseases due to GATA-3 deficiency – such as in hypoparathyroidism, sensorineural deafness, and renal (HDR) syndrome – by OPG therapy.
Collapse
Affiliation(s)
- Shyan-Yuan Kao
- Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary
| | - Konstantina M Stankovic
- 1] Eaton Peabody Laboratories and Department of Otolaryngology, Massachusetts Eye and Ear Infirmary [2] Department of Otology and Laryngology, and Program in Speech and Hearing Bioscience and Technology, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Joshi S, Clapp WL, Wang W, Khan SR. Osteogenic changes in kidneys of hyperoxaluric rats. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2000-12. [PMID: 26122267 DOI: 10.1016/j.bbadis.2015.06.020] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Revised: 05/30/2015] [Accepted: 06/25/2015] [Indexed: 02/06/2023]
Abstract
Many calcium oxalate (CaOx) kidney stones develop attached to renal papillary sub-epithelial deposits of calcium phosphate (CaP), called Randall's plaque (RP). Pathogenesis of the plaques is not fully understood. We hypothesize that abnormal urinary environment in stone forming kidneys leads to epithelial cells losing their identity and becoming osteogenic. To test our hypothesis male rats were made hyperoxaluric by administration of hydroxy-l-proline (HLP). After 28days, rat kidneys were extracted. We performed genome wide analyses of differentially expressed genes and determined changes consistent with dedifferentiation of epithelial cells into osteogenic phenotype. Selected molecules were further analyzed using quantitative-PCR and immunohistochemistry. Genes for runt related transcription factors (RUNX1 and 2), zinc finger protein Osterix, bone morphogenetic proteins (BMP2 and 7), bone morphogenetic protein receptor (BMPR2), collagen, osteocalcin, osteonectin, osteopontin (OPN), matrix-gla-protein (MGP), osteoprotegrin (OPG), cadherins, fibronectin (FN) and vimentin (VIM) were upregulated while those for alkaline phosphatase (ALP) and cytokeratins 10 and 18 were downregulated. In conclusion, epithelial cells of hyperoxaluric kidneys acquire a number of osteoblastic features but without CaP deposition, perhaps a result of downregulation of ALP and upregulation of OPN and MGP. Plaque formation may additionally require localized increases in calcium and phosphate and decrease in mineralization inhibitory potential.
Collapse
Affiliation(s)
- Sunil Joshi
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - William L Clapp
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Wei Wang
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States
| | - Saeed R Khan
- Department of Pathology, Immunology and Laboratory Medicine, College of Medicine, University of Florida, Gainesville, FL, United States; Department of Urology, College of Medicine, University of Florida, Gainesville, FL, United States.
| |
Collapse
|
13
|
Anti-osteoporotic activity of sialoglycoproteins isolated from the eggs of Carassius auratus by promoting osteogenesis and increasing OPG/RANKL ratio. J Funct Foods 2015. [DOI: 10.1016/j.jff.2015.03.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
14
|
Wyganowska-Świątkowska M, Urbaniak P, Nohawica MM, Kotwicka M, Jankun J. Enamel matrix proteins exhibit growth factor activity: A review of evidence at the cellular and molecular levels. Exp Ther Med 2015; 9:2025-2033. [PMID: 26161150 DOI: 10.3892/etm.2015.2414] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/25/2015] [Indexed: 01/23/2023] Open
Abstract
Enamel matrix derivative (EMD) is a commercially available protein extract, mainly comprising amelogenins. A number of other polypeptides have been identified in EMD, mostly growth factors, which promote cementogenesis and osteogenesis during the regeneration processes through the regulation of cell proliferation, differentiation and activity; however, not all of their functions are clear. Enamel extracts have been proposed to have numerous activities such as bone morphogenetic protein- and transforming growth factor β (TGF-β)-like activity, and activities similar to those of insulin-like growth factor, fibroblast growth factor, platelet-derived growth factor, vascular endothelial growth factor and epidermal growth factor. These activities have been observed at the molecular and cellular levels and in numerous animal models. Furthermore, it has been suggested that EMD contains an unidentified biologically active factor that acts in combination with TGF-β1, and several studies have reported functional similarities between growth factors and TGF-β in cellular processes. The effects of enamel extracts on the cell cycle and biology are summarized and discussed in this review.
Collapse
Affiliation(s)
| | - Paulina Urbaniak
- Department of Cell Biology, Poznan University of Medical Sciences, Poznań 60-806, Poland
| | | | - Małgorzata Kotwicka
- Department of Cell Biology, Poznan University of Medical Sciences, Poznań 60-806, Poland
| | - Jerzy Jankun
- Department of Urology, Urology Research Centre, College of Medicine, University of Toledo, Toledo, OH 43614, USA ; Protein Research Chair, Department of Biochemistry, College of Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia ; Department of Clinical Nutrition, Medical University of Gdańsk, Gdańsk 80-211, Poland
| |
Collapse
|
15
|
Sung YY, Lee CS. Mammary gland-specific expression of biologically active human osteoprotegerin in transgenic mice. Dev Reprod 2013; 17:1-8. [PMID: 25949115 PMCID: PMC4282218 DOI: 10.12717/dr.2013.17.1.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2012] [Revised: 01/28/2013] [Accepted: 02/09/2013] [Indexed: 12/05/2022]
Abstract
Osteoprotegerin (OPG) is a secreted glycoprotein that regulates bone resorption by inhibiting differentiation and activation of osteoclast, thereby potentially useful for the treatment of many bone diseases associated with increased bone loss. In this study, we designed a novel cDNA expression cassette by modifying the potent and mammary gland-specific goat β-casein/hGH hybrid gene construct and examined human OPG (hOPG) cDNA expression in transgenic mice. Six transgenic mice all successfully expressed hOPG in their milk at the level of 0.06-2,000 µg/ml. An estimated molecular weight of the milk hOPG was 55 kDa in SDS-PAGE, which is the same as a naturally glycosylated monomer. This hOPG expression was highly specific to the mammary glands of transgenic mice. hOPG mRNA was not detected in any organs analyzed except mammary gland. Functional integrity of milk hOPG was evaluated by TRAP (tartrate-resistant acid phosphatase) activity assay in bone marrow cell cultures. OPG ligand (OPG-L) treatment increased TRAP activity by two fold but it was completely abolished by co-treatment with transgenic milk containing hOPG. Taken together, our novel cDNA expression cassette could direct an efficient expression of biologically active hOPG, a potential candidate pharmaceutical for bone diseases, only in the mammary gland of transgenic mice.
Collapse
Affiliation(s)
- Yoon-Young Sung
- Basic Herbal Medicine Research Group, Korea Institute of Oriental Medicine, Daejeon 305-811, Korea
| | - Chul-Sang Lee
- Department of Biology, Kunsan National University, Kunsan 573-701, Korea
| |
Collapse
|
16
|
Szpalski C, Sagebin F, Barbaro M, Warren SM. The influence of environmental factors on bone tissue engineering. J Biomed Mater Res B Appl Biomater 2012; 101:663-75. [PMID: 23165885 DOI: 10.1002/jbm.b.32849] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2012] [Revised: 09/28/2012] [Accepted: 10/05/2012] [Indexed: 12/14/2022]
Abstract
Bone repair and regeneration are dynamic processes that involve a complex interplay between the substrate, local and systemic cells, and the milieu. Although each constituent plays an integral role in faithfully recreating the skeleton, investigators have long focused their efforts on scaffold materials and design, cytokine and hormone administration, and cell-based therapies. Only recently have the intangible aspects of the milieu received their due attention. In this review, we highlight the important influence of environmental factors on bone tissue engineering.
Collapse
Affiliation(s)
- Caroline Szpalski
- Department of Plastic Surgery, New York University Langone Medical Center, New York, New York, USA
| | | | | | | |
Collapse
|
17
|
Wang W, Li F, Wang K, Cheng B, Guo X. PAPSS2 promotes alkaline phosphates activity and mineralization of osteoblastic MC3T3-E1 cells by crosstalk and Smads signal pathways. PLoS One 2012; 7:e43475. [PMID: 22916269 PMCID: PMC3420899 DOI: 10.1371/journal.pone.0043475] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 07/20/2012] [Indexed: 12/17/2022] Open
Abstract
Several studies have indicated that PAPSS2 (3'-phosphoadenosine-5'-phosphosulfate synthetase 2) activity is important to normal skeletal development. Mouse PAPSS2 is predominantly expressed during the formation of the skeleton and cartilaginous elements of the mouse embryo and in newborn mice. However, the role and mechanism of PAPSS2 in bone formation remains largely unidentified. By analyzing the expression pattern of the PAPSS2 gene, we have found that PAPSS2 is expressed in bone tissue and bone formation. PAPSS2 transcripts increase during osteoblast differentiation and are in less level in RANKL-induced osteoclast like cells. By using lentivirus-mediated RNA interference (RNAi) technology, we knocked down PAPSS2 expression in MC3T3-E1 osteoblast. Silencing of PAPSS2 expression significantly decreases ALP activity and cell mineralization, inhibits expression of osteoblast marker osteopontin (OPN) and collagen I. Conversely, overexpression of PAPSS2 promotes the MC3T3-E1 to differentiate into osteoblast and mineralization. Moreover, compared to that in the control cells, the mRNA level and protein expression of phosphorylated Smad 2/3, which is a key transcriptional factor in the Smad osteoblast differentiation pathway, showed significant decreases in PAPSS2-silenced cells and increases in PAPSS2-overexpression cells. These results suggest that PAPSS2 might regulate osteoblast ALP activity and cell mineralization, probably through Smads signal pathways.
Collapse
Affiliation(s)
- Weizhuo Wang
- Department of Orthopedics Surgery, the Second Affiliated Hospital, College of Medicine, Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China.
| | | | | | | | | |
Collapse
|
18
|
de Freitas A, Banerjee S, Xie N, Cui H, Davis KI, Friggeri A, Fu M, Abraham E, Liu G. Identification of TLT2 as an engulfment receptor for apoptotic cells. THE JOURNAL OF IMMUNOLOGY 2012; 188:6381-8. [PMID: 22573805 DOI: 10.4049/jimmunol.1200020] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Clearance of apoptotic cells (efferocytosis) is critical to the homeostasis of the immune system by restraining inflammation and autoimmune response to intracellular Ags released from dying cells. TLRs-mediated innate immunity plays an important role in pathogen clearance and in regulation of the adaptive immune response. However, the regulation of efferocytosis by activation of TLRs has not been well characterized. In this study, we found that activation of TLR3 or TLR9, but not of TLR2, enhances engulfment of apoptotic cells by macrophages. We found that the activation of TLR3 upregulates the expression of triggering receptor expressed on myeloid cells (TREM)-like protein 2 (TLT2), a member of the TREM receptor family, on the surface of macrophages. Blocking TLT2 on the macrophage surface by either specific anti-TLT2 Ab or soluble TLT2 extracellular domain attenuated the enhanced ability of macrophages with TLR3 activation to engulf apoptotic cells. To the contrary, overexpression of TLT2 increased the phagocytosis of apoptotic cells. We found that TLT2 specifically binds to phosphatidylserine, a major "eat me" signal that is exposed on the surface of apoptotic cells. Furthermore, we found that TLT2 mediates phagocytosis of apoptotic cells in vivo. Thus, our studies identified TLT2 as an engulfment receptor for apoptotic cells. Our data also suggest a novel mechanism by which TREM receptors regulate inflammation and autoimmune response.
Collapse
Affiliation(s)
- Andressa de Freitas
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Shah NJ, Hong J, Hyder MN, Hammond PT. Osteophilic multilayer coatings for accelerated bone tissue growth. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2012; 24:1445-1450. [PMID: 22311551 PMCID: PMC3870474 DOI: 10.1002/adma.201104475] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2011] [Revised: 12/30/2011] [Indexed: 05/27/2023]
Abstract
Osteophilic modular nanostructured multilayers containing hydroxyapatite nanoparticles complexed with a natural polymer chitosan create an osteoconductive surface for mesenchymal stem cells (MSCs). Coupled with the sustained release of physiological amounts of osteoinductive bone morphogenetic protein over several days from degradable poly(β-amino ester) based multilayers, this single coating results in a synergistic accelerated and upregulated differentiation of MSCs into osteoblasts laying down new bone tissue on orthopedic implants.
Collapse
|
20
|
Gradosova I, Zivna H, Palicka V, Hubena S, Svejkovska K, Zivny P. Protective effect of amlodipine on rat bone tissue after orchidectomy. Pharmacology 2012; 89:37-43. [PMID: 22302040 DOI: 10.1159/000335491] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 12/01/2011] [Indexed: 11/19/2022]
Abstract
AIM Our study aimed to investigate the effect of amlodipine on bone metabolism in orchidectomized rats. METHODS Eight-week-old rats were divided into three groups. The sham-operated control group (SHAM) and the control group after orchidectomy (ORX) received the standard laboratory diet (SLD). The experimental group after orchidectomy (ORX+AML) received SLD enriched with amlodipine for 12 weeks. Bone marker concentrations in serum of PINP, OPG and IGF-1, and the levels of CTX-I, BAP and BMP-2 in a bone homogenate were measured using enzyme-linked immunosorbent assay. Bone mineral density (BMD) was measured by dual energy X-ray absorptiometry. The femurs were used for biomechanical testing. RESULTS Bone markers (CTX-I, BAP, BMP-2) in ORX were higher versus SHAM. In ORX+AML there was a decrease in PINP, CTX-I, BAP, BMP-2 and OPG versus ORX. IGF-1 was decreased in ORX versus SHAM. In ORX+AML it was increased versus ORX. In ORX, a decrease was demonstrated versus SHAM in BMD of the whole body, in the lumbar vertebrae and in both femurs. In ORX+AML there was an increase in BMD of the whole body versus ORX. Three-point bending test revealed a decrease in maximal load values in ORX versus SHAM. After amlodipine administration there was an increase in the left femur versus ORX. CONCLUSIONS Amlodipine is capable of mitigating the negative effects of orchidectomy and could be a good prevention of osteoporosis.
Collapse
Affiliation(s)
- Iveta Gradosova
- Institute of Clinical Biochemistry and Diagnostics, Charles University in Prague, Faculty of Medicine and University Hospital in Hradec Kralove, Kralove, Czech Republic. gradosovai@ lfhk.cuni.cz
| | | | | | | | | | | |
Collapse
|
21
|
Banerjee S, de Freitas A, Friggeri A, Zmijewski JW, Liu G, Abraham E. Intracellular HMGB1 negatively regulates efferocytosis. THE JOURNAL OF IMMUNOLOGY 2011; 187:4686-94. [PMID: 21957148 DOI: 10.4049/jimmunol.1101500] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
High mobility group box 1 (HMGB1) is a highly conserved protein with multiple intracellular and extracellular functions, including transcriptional regulation, as well as modulation of inflammation, cell migration, and ingestion of apoptotic cells. In these experiments, we examined a potential role for intracellular HMGB1 in modulating phagocytosis. We found that phagocytosis of apoptotic cells resulted in translocation of HMGB1 into the cytoplasm and extracellular space. Transient or stable inhibition of HMGB1 expression in bone marrow-derived macrophages or fibroblasts resulted in increased phagocytosis of apoptotic thymocytes and apoptotic neutrophils. Knockdown of HMGB1 was associated with enhanced activation of Rac-1 and cytoskeletal rearrangement. Intracellular events involved in phagocytosis and upstream of Rac-1 activation, such as phosphorylation of ERK and focal adhesion kinase (FAK), were increased after knockdown of HMGB1. Inhibition of Src kinase activity prevented the increase in phosphorylation of FAK and ERK present during phagocytosis in HMGB1 knockdown cells, and also abrogated the enhancement in phagocytosis associated with HMGB1 knockdown. Interaction between Src and FAK in the cytoplasm of HMGB1 knockdown fibroblasts was enhanced compared with that present in control fibroblasts. Under in vitro conditions, the presence of HMGB1 diminished interactions between purified FAK and Src. These studies demonstrate a novel role for HMGB1 in the regulation of phagocytosis. In particular, these experiments show that intracellular HMGB1, through associating with Src kinase and inhibiting interactions between Src and FAK, diminishes the phagocytic ability of macrophages and other cell populations.
Collapse
Affiliation(s)
- Sami Banerjee
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | | | | | | | | | |
Collapse
|
22
|
Rath B, Nam J, Deschner J, Schaumburger J, Tingart M, Grässel S, Grifka J, Agarwal S. Biomechanical forces exert anabolic effects on osteoblasts by activation of SMAD 1/5/8 through type 1 BMP receptor. Biorheology 2011; 48:37-48. [PMID: 21515935 DOI: 10.3233/bir-2011-0580] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Osteoblasts are mechanosensitive cells, which respond to biomechanical stimuli to regulate the bone structure through anabolic and catabolic gene regulation. To examine the effects of mechanical forces on the osteogenic responses through the SMAD signaling in osteoblasts, the cells were cultured in well-characterized mechanoresponsive 3-D scaffolds and exposed to 10% dynamic compressive strain (Cmp) at 1 Hz. Subsequently, SMAD phosphorylation and osteogenic gene induction was examined. Osteoblasts cultured in 3-D scaffolds exhibited increased constitutive SMAD 1/5/8 phosphorylation, as compared to monolayers cultures. This SMAD 1/5/8 phosphorylation was further upregulated after 10, 30 and 60 min in response to Cmp, exhibiting a peak activation at 30 min. No significant changes in SMAD2 phosphorylation were observed, suggesting signals generated by Cmp may not activate the Transforming Growth Factor-β signaling cascade. Subsequently, biomechanical stimulation-induced SMAD 1/5/8 phosphorylation upregulated the expression of osteogenic genes such as Osteoprotegrin, Msx2 and Runx2. Dorsomorphin, a selective inhibitor of the bone morphogenetic protein (BMP) receptor type 1 (BMPR1), blocked Cmp-induced SMAD 1/5/8 phosphorylation, as well as Osteoprotegrin, Msx2 and Runx2 gene expression. Collectively, the present findings demonstrate that biomechanical stimulation of osteoblasts activates SMAD 1/5/8 in the BMP signaling pathway through BMPR1 and may enhance osteogenesis by upregulating SMAD-dependent osteogenic genes.
Collapse
Affiliation(s)
- B Rath
- Department of Orthopaedic Surgery, University of Regensburg, Bad Abbach, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Adwan H, Zhivkova-Galunska M, Georges R, Eyol E, Kleeff J, Giese NA, Friess H, Bergmann F, Berger MR. Expression of HOXC8 is inversely related to the progression and metastasis of pancreatic ductal adenocarcinoma. Br J Cancer 2011; 105:288-95. [PMID: 21712827 PMCID: PMC3142801 DOI: 10.1038/bjc.2011.217] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background: The transcription factor HOXC8 regulates many genes involved in tumour progression. This study was to investigate the role of HOXC8 in pancreatic ductal adenocarcinoma (PDAC) growth and metastasis. Methods: The Hoxc8 expression was determined in 15 PDAC cell lines and human specimens by RT–polymerase chain reaction and/or immunohistochemistry. The effects of HOXC8 silencing by RNA interference were investigated by functional tests. Results: The Hoxc8 mRNA expression in PDAC cell lines was negatively related to their growth in vivo. Except for Suit2-007 cells, only those with low Hoxc8 mRNA expression grew in nude rats. Successful down-regulation of HOXC8 expression caused increased proliferation, migration (P⩽0.05) and colony formation (P⩽0.05) in Suit2-007, Panc-1 and MIA PaCa-2 PDAC cells, respectively. The Hoxc8 mRNA levels in diseased human pancreas tissues were significantly increased over normal in PDAC and autoimmune chronic pancreatitis specimens (P<0.01, respectively), but negatively related to tumour stage (P=0.09). In primary and metastatic tumour samples, immunohistochemical staining for HOXC8 was stronger in surrounding than in neoplastic tissues. Furthermore, grading of primary carcinomas was negatively associated with HOXC8 staining (P=0.03). Liver metastases showed the lowest HOXC8 expression of all neoplastic lesions. Conclusion: These data indicate that HOXC8 expression is inversely related to PDAC progression and metastasis.
Collapse
Affiliation(s)
- H Adwan
- Toxicology and Chemotherapy Unit, German Cancer Research Center, Deutsches Krebsforschungszentrum Heidelberg, Im Neuenheimer Feld 581, Heidelberg D-69120, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Walrad PB, Hang S, Gergen JP. Hairless is a cofactor for Runt-dependent transcriptional regulation. Mol Biol Cell 2011; 22:1364-74. [PMID: 21325629 PMCID: PMC3078061 DOI: 10.1091/mbc.e10-06-0483] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Runt is a vital transcriptional regulator in the developmental pathway responsible for segmentation in the Drosophila embryo. Runt activates or represses transcription in a manner that is dependent on both cellular context and the specific downstream target. Here we identify Hairless (H) as a Runt-interacting molecule that functions during segmentation. We find that H is important for maintenance of engrailed (en) repression as was previously demonstrated for Groucho (Gro), Rpd3, and CtBP. H also contributes to the Runt-dependent repression of sloppy-paired-1 (slp1), a role that is not shared with these other corepressors. We further find distinct roles for these different corepressors in the regulation of other Runt targets in the early Drosophila embryo. These findings, coupled with observations on the distinct functional requirements for Runt in regulating these several different targets, indicate that Runt-dependent regulation in the Drosophila blastoderm embryo relies on unique, target-gene-specific molecular interactions.
Collapse
Affiliation(s)
- Pegine B Walrad
- Department of Biochemistry and Cell Biology, Stony Brook University, Stony Brook, NY 11794, USA
| | | | | |
Collapse
|
25
|
Sethi N, Kang Y. Dysregulation of developmental pathways in bone metastasis. Bone 2011; 48:16-22. [PMID: 20630490 DOI: 10.1016/j.bone.2010.07.005] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 06/30/2010] [Accepted: 07/06/2010] [Indexed: 02/07/2023]
Abstract
It is well-known that pathways normally functioning during embryonic development are dysregulated in cancer. Experimental and clinical studies have established strong connections between aberrant developmental pathways and transformation, as well as other early stage events of cancer progression. There is now emerging evidence that also indicates the contribution of developmental pathways to the pathogenesis of distant metastasis, including bone metastasis. In particular, the Wnt, BMP, and Hedgehog signaling pathways have all been implicated in the development of bone metastasis. These developmental pathways participate in the regulation of cell-autonomous functions in tumor cells as well as tumor-stromal interactions in the bone microenvironment, eventually promoting the formation of osteolytic or osteoblastic bone metastasis.
Collapse
Affiliation(s)
- Nilay Sethi
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | | |
Collapse
|
26
|
Abstract
Smad proteins are intracellular molecules that mediate the canonical signaling cascade of TGFbeta superfamily growth factors. The TGFbeta superfamily comprises two groups of growth factors, BMPs and TGFbetas. Both groups can be further divided into several sub-groups based on sequence homologies and functional similarities. Ligands of the TGFbeta superfamily bind to cell surface receptors to activate Smad proteins in the cytoplasm; then the activated Smad proteins translocate into the nucleus to activate or repress specific target gene transcription. Both groups of growth factors play important roles in skeletal development and regeneration. However, whether these effects reflect signaling through canonical Smad pathways, or other non-canonical signaling pathways in vivo remains a mystery. Moreover, the mechanisms utilized by Smad proteins to initiate nuclear events and their interactions with cytoplasmic proteins are still under intensive investigation. This review will discuss the most recent progress understanding Smad signaling in the context of skeletal development and regeneration.
Collapse
Affiliation(s)
- Buer Song
- Orthopedic Hospital Research Center, Department of Orthopedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, United States
| | | | | |
Collapse
|
27
|
Allori AC, Sailon AM, Warren SM. Biological basis of bone formation, remodeling, and repair-part I: biochemical signaling molecules. TISSUE ENGINEERING PART B-REVIEWS 2009; 14:259-73. [PMID: 18665803 DOI: 10.1089/ten.teb.2008.0082] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
The bony biochemical environment is an active and dynamic system that permits and promotes cellular functions that lead to matrix production and ossification. Each component is capable of conveying important regulatory cues to nearby cells, thus effecting gene expression and changes at the cytostructural level. Here, we review the various signaling molecules that contribute to the active and dynamic nature of the biochemical system. These components include hormones, cytokines, and growth factors. We describe their role in regulating bone metabolism. Certain growth factors (i.e., TGF-beta, IGF-1, and VEGF) are described in greater detail because of their potential importance in developing successful tissue-engineering strategies.
Collapse
Affiliation(s)
- Alexander C Allori
- Institute of Reconstructive Plastic Surgery, New York University Medical Center, New York, New York, USA
| | | | | |
Collapse
|
28
|
Bone morphogenetic protein 2 enhances PGE(2)-stimulated osteoclast formation in murine bone marrow cultures. Prostaglandins Other Lipid Mediat 2009; 90:76-80. [PMID: 19744575 DOI: 10.1016/j.prostaglandins.2009.08.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2009] [Revised: 08/27/2009] [Accepted: 08/28/2009] [Indexed: 01/16/2023]
Abstract
Bone morphogenetic protein 2 (BMP-2) is used clinically to stimulate bone formation and accelerate fracture repair. Adding prostaglandin (PG) E(2) or PGE(2) receptor agonists to BMP-2 has been proposed to improve BMP-2 efficacy. However, this may enhance bone resorption, since PGE(2) can increase receptor activator of NF-kappaB ligand (RANKL) expression and decrease osteoprotegerin (OPG) expression in osteoblasts, and the RANKL:OPG ratio is critical for osteoclast formation. We used bone marrow (BM) cultures and BM macrophage (BMM) cultures from outbred CD1 mice to examine effects on osteoclast formation of BMP-2 and PGE(2). In BM cultures, which contain both osteoblastic and osteoclastic lineage cells, BMP-2 (100 ng/ml) alone did not increase osteoclast formation but enhanced the peak response to PGE(2) by 1.6-9.6-fold. In BMM cultures, which must be treated with RANKL because they do not contain osteoblastic cells, BMP-2 did not increase osteoclast formation, with or without PGE(2). Our results suggest that BMP-2 can increase osteoclast formation in response to PGE(2) by increasing the RANKL:OPG ratio in osteoblasts, which may have therapeutic implications for the use of BMP-2.
Collapse
|
29
|
Deng H, Ravikumar T, Yang WL. Overexpression of bone morphogenetic protein 4 enhances the invasiveness of Smad4-deficient human colorectal cancer cells. Cancer Lett 2009; 281:220-31. [DOI: 10.1016/j.canlet.2009.02.046] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Revised: 02/19/2009] [Accepted: 02/24/2009] [Indexed: 12/15/2022]
|
30
|
Tyrovola JB, Spyropoulos MN, Makou M, Perrea D. Root resorption and the OPG/RANKL/RANK system: a mini review. J Oral Sci 2009; 50:367-76. [PMID: 19106463 DOI: 10.2334/josnusd.50.367] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022]
Abstract
Odontoclastic root resorption is a significant clinical issue in relation to orthodontic tooth movement, and resorption of the roots of primary teeth is an intriguing biological phenomenon. The functional coordination of the OPG/RANKL/RANK system seems to contribute not only to alveolar remodeling, but also to resorption during orthodontic tooth movement and physiological root resorption. Serum OPG and s-RANKL are related to regulation of bone homeostasis by the OPG/RANKL/RANK system, and determination of their concentrations might be useful for predicting the rate of bone remodeling during orthodontic tooth movement, the net effect between bone remodeling and root resorption, and the degree of root resorption. It is therefore rational to speculate that a study of the levels of OPG and s-RANKL in blood and GCF, in relation to the degree of root resorption during orthodontic tooth movement, using healthy experimental animals and a carefully planned and organized experimental design, may be able to answer this intriguing question.
Collapse
Affiliation(s)
- Joanna B Tyrovola
- Department of Orthodontics, School of Dentistry, University of Athens, Athens, Greece.
| | | | | | | |
Collapse
|
31
|
Zheng YJ, Chung HJ, Min H, Kang M, Kim SH, Gadi J, Kim MH. In vitro osteoblast differentiation is negatively regulated by Hoxc8. Appl Biochem Biotechnol 2009; 160:891-900. [PMID: 19214787 DOI: 10.1007/s12010-009-8558-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2008] [Accepted: 02/03/2009] [Indexed: 10/21/2022]
Abstract
Hoxc8 has multiple roles in normal skeletal development. In this paper, a MC3T3-E1 subclone 4 osteogenic cell differentiation model was used to examine expression of Hoxc8 at multiple stages of osteogenesis. We found that Hoxc8 expression levels do not change in the early stage but increase in the middle stage and decrease in the late stage of osteogenesis. A knockdown of Hoxc8 by small-interfering RNA transfection in C2C12 cells indicated that Hoxc8 is a negative regulator of osteogenesis. Similarly, expression of Hoxc8 in C2C12 cells decreases alkaline phosphatase levels induced by bone morphogenetic protein-2 (BMP-2). The results of this study showed that Hoxc8 is involved in BMP-2-induced osteogenesis, and osteoblast differentiation in vitro is negatively regulated by Hoxc8, suggesting that Hoxc8 regulation is essential for osteoblast differentiation.
Collapse
Affiliation(s)
- Yan-Jun Zheng
- Department of Anatomy, Embryology Lab, Brain Korea 21 Project for Medical Science, College of Medicine, Yonsei University, 134 Seodaemun-gu, Shinchon-dong, 120-752 Seoul, South Korea
| | | | | | | | | | | | | |
Collapse
|
32
|
Sato MM, Nakashima A, Nashimoto M, Yawaka Y, Tamura M. Bone morphogenetic protein-2 enhances Wnt/β-catenin signaling-induced osteoprotegerin expression. Genes Cells 2009; 14:141-53. [DOI: 10.1111/j.1365-2443.2008.01258.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
33
|
El Hadj Othmane T, Speer G, Fekete B, Szabó T, Egresits J, Fodor E, Kiss I, Nemcsik J, Szabó A, Németh Z, Szathmári M, Tislér A. [Osteoprotegerin: regulator, protector and marker]. Orv Hetil 2008; 149:1971-80. [PMID: 18842549 DOI: 10.1556/oh.2008.28470] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Experimental and clinical trials in the field of bone biology helped to clarify the role of receptors, which belong to the tumor necrosis factor family, such as osteoprotegerin and receptor activator of nuclear factor kappaB (RANK), in the regulation of bone remodeling. The ligand of the receptor activator of nuclear factor kappaB (RANKL) is a stimulator of bone resorption, while osteoprotegerin is the soluble "decoy" receptor to RANKL, protecting thereby bone from resorption. Pathological states of bone remodeling (like osteoporosis) are associated with imbalance in the activity of osteoprotegerin and the receptor activator of nuclear factor kappaB. Recent studies, however, also indicate that the osteoprotegerin/RANKL/RANK system has important roles in the regulation of the immune and vascular system as well. In this review we summarize the function and regulation of osteoprotegerin, its role in pathological states--primarily in cardiovascular diseases--and its relevance as a marker of cardiovascular risk. Finally, we present our prospective trial performed among the chronic dialyzed patients, where we examined the association between the cardiovascular mortality, osteoprotegerin levels and the arterial stiffness.
Collapse
Affiliation(s)
- Taha El Hadj Othmane
- Semmelweis Egyetem, Altalános Orvostudományi Kar, I. Belgyógyászati Klinika, Budapest.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Dai J, Hall CL, Escara-Wilke J, Mizokami A, Keller JM, Keller ET. Prostate cancer induces bone metastasis through Wnt-induced bone morphogenetic protein-dependent and independent mechanisms. Cancer Res 2008; 68:5785-94. [PMID: 18632632 DOI: 10.1158/0008-5472.can-07-6541] [Citation(s) in RCA: 113] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Prostate cancer (PCa) is frequently accompanied by osteosclerotic (i.e., excessive bone production) bone metastases. Although bone morphogenetic proteins (BMP) and Wnts are mediators of PCa-induced osteoblastic activity, the relation between them in PCa bone metastases is unknown. The goal of this study was to define this relationship. Wnt3a and Wnt5a administration or knockdown of DKK-1, a Wnt inhibitor, induced BMP-4 and 6 expression and promoter activation in PCa cells. DKK-1 blocked Wnt activation of the BMP promoters. Transfection of C4-2B cells with axin, an inhibitor of canonical Wnt signaling, blocked Wnt3a but not Wnt5a induction of the BMP promoters. In contrast, Jnk inhibitor I blocked Wnt5a but not Wnt3a induction of the BMP promoters. Wnt3a, Wnt5a, and conditioned medium (CM) from C4-2B or LuCaP23.1 cells induced osteoblast differentiation in vitro. The addition of DKK-1 and Noggin, a BMP inhibitor, to CM diminished PCa CM-induced osteoblast differentiation in a synergistic fashion. However, pretreatment of PCa cells with DKK-1 before collecting CM blocked osteoblast differentiation, whereas pretreatment with Noggin only partially reduced osteoblast differentiation, and pretreatment with both DKK-1 and Noggin had no greater effect than pretreatment with DKK-1 alone. Additionally, knockdown of BMP expression in C4-2B cells inhibited Wnt-induced osteoblastic activity. These results show that PCa promotes osteoblast differentiation through canonical and noncanonical Wnt signaling pathways that stimulate both BMP-dependent and BMP-independent osteoblast differentiation. These results show a clear link between Wnts and BMPs in PCa-induced osteoblast differentiation and provide novel targets, including the noncanonical Wnt pathway, for therapy of PCa.
Collapse
Affiliation(s)
- Jinlu Dai
- Department of Urology, University of Michigan Health System, Ann Arbor, Michigan 48109-0940, USA
| | | | | | | | | | | |
Collapse
|
35
|
Susperregui ARG, Viñals F, Ho PWM, Gillespie MT, Martin TJ, Ventura F. BMP-2 regulation of PTHrP and osteoclastogenic factors during osteoblast differentiation of C2C12 cells. J Cell Physiol 2008; 216:144-52. [PMID: 18247361 DOI: 10.1002/jcp.21389] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Bone morphogenetic protein-2 (BMP-2) is strongly involved in the induction of osteoblast differentiation from mesenchymal cell precursors, as well as in enhancing bone matrix production by osteoblastic cells. Likewise, the osteoporotic phenotype of PTHrP deficient mice makes clear the importance of this paracrine regulator in bone physiology. Here, we report that BMP-2 rapidly down-regulated PTHrP gene expression through a transcriptional mechanism in pluripotent mesenchymal C2C12 cells, whereas BMP-2 increased expression of PTHrP receptor. PTHrP did not significantly alter the BMP-dependent Smad transcriptional pathway. Similarly, PTHrP did not significantly modify the BMP-regulated expression of RANKL or OPG, cytokines involved in osteoclastogenesis. More importantly, addition of PTHrP, through the PKA signaling pathway, partially prevented the BMP-dependent induction of some osteogenic markers such as Runx2 and Osterix in C2C12 cells. Our data suggest that BMP-2 down-regulation of PTHrP could facilitate terminal differentiation of osteoblasts.
Collapse
Affiliation(s)
- Antonio R G Susperregui
- IDIBELL, Departament de Ciències Fisiològiques II, Universitat de Barcelona, L'Hospitalet de Llobregat, Spain
| | | | | | | | | | | |
Collapse
|
36
|
Wallin R, Schurgers L, Wajih N. Effects of the blood coagulation vitamin K as an inhibitor of arterial calcification. Thromb Res 2008; 122:411-7. [PMID: 18234293 PMCID: PMC2529147 DOI: 10.1016/j.thromres.2007.12.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 11/01/2007] [Accepted: 12/06/2007] [Indexed: 10/22/2022]
Abstract
INTRODUCTION The transformation of smooth muscle cells (VSMCs) in the vessel wall to osteoblast like cells is known to precede arterial calcification which may cause bleeding complications. The vitamin K-dependent protein MGP has been identified as an inhibitor of this process by binding BMP-2, a growth factor known to trigger the transformation. In this study, we determined if the vitamin K-dependent Gla region in MGP by itself can inhibit the growth factor activity of BMP-2 and if menaquinone-4 (MK4) regulates gene expression in VSMCs. MATERIALS AND METHODS A synthetic gamma-carboxyglutamic acid (Gla) containing peptide covering the Gla region in human MGP was used to test its ability to inhibit BMP-2 induced transformation of mouse pro-myoblast C2C12 cells into osteoblasts. MK4 was tested by microarray analysis as a gene regulatory molecule in VSMCs. RESULTS AND CONCLUSIONS The results show that the Gla - but not the Glu-peptide inhibited the transformation which provide evidence that the Gla region in MGP is directly involved in the BMP-2/MGP interaction and emphasizes the importance of the vitamin K-dependent modification of MGP. From the data obtained from the microarray analysis, we focused on two quantitatively altered cDNAs representing proteins known to be associated with vessel wall calcification. DT-diaphorase of the vitamin K-cycle, showed increased gene expression with a 4.8-fold higher specific activity in MK4 treated cells. Osteoprotegrin gene expression was down regulated and osteoprotegrin protein secretion from the MK4 treated cells was lowered to 1.8-fold. These findings suggest that MK4 acts as an anti-calcification component in the vessel wall.
Collapse
Affiliation(s)
- Reidar Wallin
- Department of Internal Medicine, Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, North Carolina 27157, USA.
| | | | | |
Collapse
|
37
|
Holecki M, Zahorska-Markiewicz B, Janowska J, Nieszporek T, Wojaczyńska-Stanek K, Zak-Gołab A, Wiecek A. The influence of weight loss on serum osteoprotegerin concentration in obese perimenopausal women. Obesity (Silver Spring) 2007; 15:1925-9. [PMID: 17712108 DOI: 10.1038/oby.2007.229] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To assess the influence of weight reduction therapy on serum osteoprotegerin (OPG) concentration in obese patients and compare these results with normal-weight controls. RESEARCH METHODS AND PROCEDURES Forty-three obese women (BMI, 36.7 +/- 4.1 kg/m2; mean age, 50.1 +/- 4.5 years) were studied. The control group consisted of 19 normal-weight women (BMI, 24.2 +/- 2.1 kg/m2; mean age, 53.8 +/- 5.2 years). In all patients, serum concentrations of OPG, C telopeptide of type I collagen containing the cross-linking site (CTX), osteocalcin, parathormone, 25-(OH)-D3 (vitamin D), and total calcium and phosphorus were assessed before and after a 3-month weight reduction therapy. RESULTS In obese subjects, serum concentrations of OPG, 25-(OH)-D3, osteocalcin, total calcium, and phosphorus were significantly lower, and serum concentration of parathormone was significantly higher, before weight reduction therapy in comparison with normal-weight controls. After weight reduction, a significantly higher serum concentration of 25-(OH)-D3 and CTX and significantly lower concentration of OPG were found. DISCUSSION Serum concentration of OPG was significantly lower in obese patients in comparison with normal-weight controls. Weight reduction therapy resulted in further decrease in OPG serum concentrations. Therefore, OPG cannot be treated as a protective factor from bone loss in obese patients.
Collapse
Affiliation(s)
- Michał Holecki
- Department of Pathophysiology, Medical University of Silesia in Katowice, ul. Medyków 18, 40-752 Katowice, Poland.
| | | | | | | | | | | | | |
Collapse
|
38
|
Massip L, Ectors F, Deprez P, Maleki M, Behets C, Lengelé B, Delahaut P, Picard J, Rezsöhazy R. Expression of Hoxa2 in cells entering chondrogenesis impairs overall cartilage development. Differentiation 2007; 75:256-67. [PMID: 17359301 DOI: 10.1111/j.1432-0436.2006.00132.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Vertebrate Hox genes act as developmental architects by patterning embryonic structures like axial skeletal elements, limbs, brainstem territories, or neural crest derivatives. While active during the patterning steps of development, these genes turn out to be down-regulated in specific differentiation programs like that leading to chondrogenesis. To investigate why chondrocyte differentiation is correlated to the silencing of a Hox gene, we generated transgenic mice allowing Cre-mediated conditional misexpression of Hoxa2 and induced this gene in Collagen 2 alpha 1-expressing cells committed to enter chondrogenesis. Persistent Hoxa2 expression in chondrogenic cells resulted in overall chondrodysplasia with delayed cartilage hypertrophy, mineralization, and ossification but without proliferation defects. The absence of skeletal patterning anomaly and the regular migration of precursor cells indicated that the condensation step of chondrogenesis was normal. In contrast, closer examination at the differentiation step showed severely impaired chondrocyte differentiation. In addition, this inhibition affected structures independently of their embryonic origin. In conclusion, for the first time here, by a cell-type specific misexpression, we precisely uncoupled the patterning function of Hoxa2 from its involvement in regulating differentiation programs per se and demonstrate that Hoxa2 displays an anti-chondrogenic activity that is distinct from its patterning function.
Collapse
Affiliation(s)
- Laurent Massip
- Developmental Genetics Unit, Université catholique de Louvain, Brussels, Belgium.
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Juan AH, Lei H, Bhargava P, Lebrun M, Ruddle FH. Multiple roles of hoxc8 in skeletal development. Ann N Y Acad Sci 2006; 1068:87-94. [PMID: 16831908 DOI: 10.1196/annals.1346.046] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We are interested in investigating the function of Hoxc8 in skeletogenesis during mouse development. Previous studies have shown that deregulation of Hoxc8 expression in the mouse leads to several skeletal defects, such as homeotic transformation in the thoracic vertebrae, abnormal development of the rib cage, and overproliferation of chondrocytes in the hypertrophic area. By deleting a crucial enhancer of Hoxc8 in vivo, we found that precise temporal expression of Hoxc8 is important for determining the correct identity of the vertebral column in early embryos. We also identified downstream targets of Hoxc8 relevant to osteoblast differentiation at later developmental stages.
Collapse
Affiliation(s)
- Aster H Juan
- Department of Molecular, Cellular and Developmental Biology, Yale University, New Haven, CT 06511, USA.
| | | | | | | | | |
Collapse
|
40
|
Li X, Nie S, Chang C, Qiu T, Cao X. Smads oppose Hox transcriptional activities. Exp Cell Res 2006; 312:854-64. [PMID: 16405960 DOI: 10.1016/j.yexcr.2005.12.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2005] [Revised: 11/28/2005] [Accepted: 12/01/2005] [Indexed: 10/25/2022]
Abstract
BMPs and Hox proteins play crucial roles in developmental processes. Beyond their mutual regulation of gene expression, little is known about the relations between their mechanisms of actions. Previously, we have shown that Hoxc8 acts as a downstream repressor in the BMP signaling pathway. Smad1 and Smad6 interact with Hoxc8 and regulate its repression activities. The Hox family contains 39 genes divided into 13 paralogs. In this report, we systemically examined the potential functions of all the paralogous Hox proteins as BMP downstream transcription factors. Representative Hox proteins from each paralog were tested. In the gel-shift assay, we found that Smad1, Smad4, and Smad6 interacted with most of the Hox proteins in ways similar to their interactions with Hoxc8. The interactions were confirmed in mammalian cells. We also examined the effects of Smads on Hox-induced transactivation. Particularly, we determined that for Hoxd10 as a transcriptional activator, both Smad1 and Smad6 opposed its activity. In addition, Smad6 also inhibited Hoxc8- and Hoxb7-induced osteoprotegerin (OPG) transactivation. Furthermore, Smad1 inhibited Hoxb4-mediated target gene Irx5 expression during early Xenopus development. Our findings suggest that Hox proteins act as general downstream DNA-binding proteins in BMP signaling cascade and their transcriptional activities are regulated by Smads.
Collapse
Affiliation(s)
- Xuelin Li
- Department of Pathology, University of Alabama at Birmingham, 1670 University Blvd., VH G003, Birmingham, AL 35294-0019, USA
| | | | | | | | | |
Collapse
|
41
|
Hori M, Sawai H, Tsuji Y, Okamura H, Koyama K. Bone morphogenetic protein-2 counterregulates interleukin-18 mRNA and protein in MC3T3-E1 mouse osteoblastic cells. Connect Tissue Res 2006; 47:124-32. [PMID: 16753805 DOI: 10.1080/03008200600685350] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Fibroblast growth factors-2 (FGF-2) and bone morphogenetic protein-2 (BMP-2) are two of the main factors that regulate differentiation of osteoblasts. Interleukin-18 (IL-18), originally cloned as an interferon gamma-inducing factor, has been reported to inhibit maturation of osteoclasts by upregulation of osteoprotegerin secreted from osteoblasts. Little is known about the functional relationship between IL-18 and the two growth factors in osteoblast differentiation. To better understand this relationship, we analyzed the effect of BMP-2 and FGF-2 on the mRNA expression levels of IL-18, as well as IL-1alpha and IL-6, in MC3T3-E1 mouse osteoblastic cells. Following this, the effects of BMP-2 on the expression of IL-18 protein and caspase-1 protein were analyzed by immunofluorescence staining. Real-time PCR and immunofluorescence staining analysis showed that FGF-2 had no effect on the expression of IL-18 mRNA and protein, but while BMP-2 reduced IL-18 mRNA levels, increased immunostaining of both IL-18 protein and caspase-1 protein was detected in BMP-2-treated MC3T3-E1 cells. Although the significance and mechanisms of this counterregulation of IL-18 mRNA and protein were not determined in this study, the increase of IL-18 protein suggested that BMP-2 may induce an active form of IL-18.
Collapse
Affiliation(s)
- Masateru Hori
- Department of Obstetrics and Gynecology, Hyogo College of Medicine, Hyogo, Japan
| | | | | | | | | |
Collapse
|
42
|
Vidal C, Brincat M, Xuereb Anastasi A. TNFRSF11B gene variants and bone mineral density in postmenopausal women in Malta. Maturitas 2005; 53:386-95. [PMID: 16343827 DOI: 10.1016/j.maturitas.2005.11.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Revised: 10/14/2005] [Accepted: 11/01/2005] [Indexed: 11/17/2022]
Abstract
UNLABELLED A number of polymorphisms in various genes have been identified and associated with bone mineral density (BMD) and with an increased risk of osteoporosis. OBJECTIVE In this study, three single nucleotide polymorphisms (SNPs) within the TNFRSF11B gene were studied for association with an increased risk of osteoporosis in postmenopausal Maltese women (n=126). METHODOLOGY Analysis was performed by PCR restriction fragment length polymorphism (RFLP) while BMD at the lumbar spine, femoral neck, Ward's triangle and trochanter was measured by DEXA. RESULTS No significant association was observed between genotypes and BMD for all polymorphisms studied within this gene. Homozygotes CC (T(950)-C) were observed to have the highest BMD at all anatomical sites although statistical significance was not reached when comparing the three genotypes. A statistical significant difference was observed in the distribution of genotype frequencies for this polymorphism between normal individuals and those that were either osteopenic or osteoporotic at one or both anatomical sites, with the TT genotype associated more frequently with low BMD. The T(950)-C and G(1181)-C polymorphisms were in strong linkage disequilibrium with each other but not with the A(163)-G polymorphism further upstream in the OPG promoter. Statistical significance was reached when constructing haplotypes, where the A-T-G haplotype was found to be more frequent in individuals with low BMD. CONCLUSIONS These results indicate the possible role of TNFRSF11B gene variants in postmenopausal bone loss in women in Malta.
Collapse
Affiliation(s)
- C Vidal
- Department of Pathology, University of Malta Medical School, Malta
| | | | | |
Collapse
|
43
|
Dai J, Keller J, Zhang J, Lu Y, Yao Z, Keller ET. Bone morphogenetic protein-6 promotes osteoblastic prostate cancer bone metastases through a dual mechanism. Cancer Res 2005; 65:8274-85. [PMID: 16166304 DOI: 10.1158/0008-5472.can-05-1891] [Citation(s) in RCA: 158] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Prostate cancer frequently metastasizes to bone where it forms osteoblastic lesions through unknown mechanisms. Bone morphogenetic proteins (BMP) are mediators of skeletal formation. Prostate cancer produces a variety of BMPs, including BMP-6. We tested the hypothesis that BMP-6 contributes to prostate cancer-induced osteosclerosis at bone metastatic sites. Prostate cancer cells and clinical tissues produced BMP-6 that increased with aggressiveness of the tumor. Prostate cancer-conditioned medium induced SMAD phosphorylation in the preosteoblast MC3T3 cells, and phosphorylation was diminished by anti-BMP-6 antibody. Prostate cancer-conditioned medium induced mineralization of MC3T3 cells, which was blocked by both the BMP inhibitor noggin and anti-BMP-6. Human fetal bones were implanted in severe combined immunodeficient mice and after 4 weeks, LuCaP 23.1 prostate cancer cells were injected both s.c. and into the bone implants. Anti-BMP-6 or isotype antibody administration was then initiated. Anti-BMP-6 reduced LuCaP 23.1-induced osteoblastic activity, but had no effect on its osteolytic activity. This was associated with increased osteoblast numbers and osteoblast activity based on bone histomorphometric evaluation. As endothelin-1 has been implicated in bone metastases, we measured serum endothelin-1 levels but found they were not different among the treatment groups. In addition to decreased bone production, anti-BMP-6 reduced intraosseous, but not s.c., tumor size. We found that BMP-2, BMP-4, BMP-6, and BMP-7 had no direct effect on prostate cancer cell growth, but BMP-2 and BMP-6 increased the in vitro invasive ability of prostate cancer cell. These data show that prostate cancer promotes osteoblastic activity through BMP-6 and that, in addition to its bone effects, suggest that BMPs promote the ability of the prostate cancer cells to invade the bone microenvironment.
Collapse
Affiliation(s)
- Jinlu Dai
- Department of Urology, School of Medicine, University of Michigan, Ann Arbor, Michigan 48109-0940, USA
| | | | | | | | | | | |
Collapse
|
44
|
Hsu YH, Niu T, Terwedow HA, Xu X, Feng Y, Li Z, Brain JD, Rosen CJ, Laird N, Xu X. Variation in genes involved in the RANKL/RANK/OPG bone remodeling pathway are associated with bone mineral density at different skeletal sites in men. Hum Genet 2005; 118:568-77. [PMID: 16249885 DOI: 10.1007/s00439-005-0062-4] [Citation(s) in RCA: 87] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2005] [Accepted: 08/23/2005] [Indexed: 01/12/2023]
Abstract
In order to assess the contribution of polymorphisms in the RANKL (TNFSF11), RANK (TNFRSF11A) and OPG (TNFRSF11B) genes to variations in bone mineral density (BMD), a population-based cohort with 1,120 extreme low hip BMD cases or extreme high hip BMD controls was genotyped on five SNPs. We further explored the associations between these genetic variations and forearm BMDs by genotyping 266 offspring and 309 available parents from 160 nuclear families. A family-based association test was used. Significantly positive associations were found for A163G polymorphisms in the promoter regions of the OPG gene, a missense substitution in exon 7 (Ala192Val) of the RANK gene and rs9594782 SNP in the 5' UTR of the RANKL gene with BMD in men only. Men with TC/CC genotypes of the rs9594782 SNP had a 2.1 times higher risk of extremely low hip BMD (P = 0.004), and lower whole body BMD (P < 0.001). Subjects with the TC genotype of the Ala192Val polymorphism had a 40% reduced risk of having extremely low hip BMD (P < 0.01), and higher whole body BMD (P < 0.01). Subjects with the GG genotype of the A163G polymorphism had a 70% reduced risk of having extremely low hip BMD (P < 0.05), and higher whole body BMD (P < 0.01). Significant gene-gene interactions were also observed among the OPG, RANK and RANKL genes. Our findings suggest that genetic variation in genes involved in the RANKL/RANK/OPG bone remodeling pathway are strongly associated with BMD at different skeletal sites in adult men, but not in women.
Collapse
Affiliation(s)
- Yi-Hsiang Hsu
- Program for Population Genetics, Harvard School of Public Health, 665 Huntington Avenue, FXB-101, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Williams TM, Williams ME, Heaton JH, Gelehrter TD, Innis JW. Group 13 HOX proteins interact with the MH2 domain of R-Smads and modulate Smad transcriptional activation functions independent of HOX DNA-binding capability. Nucleic Acids Res 2005; 33:4475-84. [PMID: 16087734 PMCID: PMC1183491 DOI: 10.1093/nar/gki761] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interactions with co-factors provide a means by which HOX proteins exert specificity. To identify candidate protein interactors of HOXA13, we created and screened an E11.5–E12.5, distal limb bud yeast two-hybrid prey library. Among the interactors, we isolated the BMP-signaling effector Smad5, which interacted with the paralogous HOXD13 but not with HOXA11 or HOXA9, revealing unique interaction capabilities of the AbdB-like HOX proteins. Using deletion mutants, we determined that the MH2 domain of Smad5 is necessary for HOXA13 interaction. This is the first report demonstrating an interaction between HOX proteins and the MH2 domain of Smad proteins. HOXA13 and HOXD13 also bind to other BMP and TGF-β/Activin-regulated Smad proteins including Smad1 and Smad2, but not Smad4. Furthermore, HOXD13 could be co-immunoprecipitated with Smad1 from cells. Expression of HOXA13, HOXD13 or a HOXD13 homeodomain mutant (HOXD13IQN>AAA) antagonized TGF-β-stimulated transcriptional activation of the pAdtrack-3TP-Lux reporter vector in Mv1Lu cells as well as the Smad3/Smad4-activated pTRS6-E1b promoter in Hep3B cells. Finally, using mammalian one-hybrid assay, we show that transcriptional activation by a GAL4/Smad3-C-terminus fusion protein is specifically inhibited by HOXA13. Our results identify a new co-factor for HOX group 13 proteins and suggest that HOX proteins may modulate Smad-mediated transcriptional activity through protein–protein interactions without the requirement for HOX monomeric DNA-binding capability.
Collapse
Affiliation(s)
| | | | - Joanne H. Heaton
- Department of Human Genetics, University of MichiganAnn Arbor, MI, USA
| | | | - Jeffrey W. Innis
- Department of Human Genetics, University of MichiganAnn Arbor, MI, USA
- Department of Pediatrics, University of MichiganAnn Arbor, MI, USA
- To whom correspondence should be addressed. Tel: +1 734 647 3817; Fax: +1 734 763 3784;
| |
Collapse
|
46
|
REGAN JOHND, WITHERSPOON DAVIDE, FOYLE DEBORAHM. Surgical repair of root and tooth perforations. ACTA ACUST UNITED AC 2005. [DOI: 10.1111/j.1601-1546.2005.00183.x] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
47
|
Abstract
Development of the vertebrate skeleton, a complex biological event that includes diverse processes such as formation of mesenchymal condensations at the sites of future skeletal elements, osteoblast and chondrocyte differentiation, and three dimensional patterning, is regulated by many growth factors. Bone morphogenetic proteins (BMPs), members of the TGF-beta superfamily, play a pivotal role in the signaling network and are involved in nearly all processes associated with skeletal morphogenesis. BMP signals are transduced from the plasma membrane receptors to the nucleus through both Smad pathway and non-Smad pathways, and regulated by many extracellular and intercellular proteins that interact with BMPs or components of the BMP signaling pathways. To gain a better understanding of the molecular mechanisms underlying the role of BMP in early skeletal development, it is necessary to elucidate the BMP signaling transduction pathways in chondrocytes and osteoblasts. The major objective of this review was to summarize BMP signaling pathways in the context of craniofacial, axial, and limb development. In particular, this discourse will focus on recent advances of the role of different ligands, receptors, Smads, and BMP regulators in osteoblast and chondrocyte differentiation during embryonic development.
Collapse
Affiliation(s)
- Mei Wan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | | |
Collapse
|
48
|
Williams TM, Williams ME, Kuick R, Misek D, McDonagh K, Hanash S, Innis JW. Candidate downstream regulated genes of HOX group 13 transcription factors with and without monomeric DNA binding capability. Dev Biol 2005; 279:462-80. [PMID: 15733672 DOI: 10.1016/j.ydbio.2004.12.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2004] [Revised: 11/24/2004] [Accepted: 12/06/2004] [Indexed: 12/24/2022]
Abstract
Hox genes encode transcription factors that regulate the morphogenesis of developing embryos. In mammals, knowledge of the genetic pathways, including the possible direct or indirect targets, regulated by HOX proteins is extremely limited. To identify the downstream genes regulated by posterior HOX proteins, we expressed HOXA13 in mouse embryonic fibroblasts lacking paralog group 13 expression using a bicistronic HOXA13/EGFP retroviral vector. Microarray analysis identified 68 genes with significant, reproducible RNA expression changes (50 activated; 18 repressed) in stable HOXA13-expressing cells. Genes with the GO annotation terms "extracellular matrix" and "basement membrane" were greatly overrepresented, and several were shown to be regulated by HOX proteins in other studies. Among the genes strongly activated by HOXA13 were Enpp2, a bifunctional enzyme known to modulate tumor and normal cell motility and which is expressed in precartilaginous condensations; Fhl1, a transcription factor implicated in muscle cell differentiation and development; and M32486, a putative integral membrane molecule expressed in the female reproductive tract. Expression differences in the HOXA13-expressing cells were confirmed for selected downstream genes using semi-quantitative RT-PCR, and in vivo coexpression with Hoxa13 in the limb interdigital mesenchyme was demonstrated for many. For two candidates, Igfbp4 and Fstl, interdigital limb bud expression was reduced in Hoxa13 mutants. To explore whether paralogous and nonparalogous HOX proteins could regulate the same genes, we created new HOX cell lines and examined the expression of selected genes identified by the HOXA13 screen. HOXD13 similarly activated/repressed 6 tested candidates, demonstrating that multiple downstream genetic pathways may be regulated by paralog HOX proteins. In contrast, HOXA9 was only able to repress expression of some gene targets. A HOXD13 mutant, HOXD13(IQN >)(AAA), incapable of monomeric DNA-binding, activated the expression of 5 HOXA13-upregulated genes; but was incapable of repressing the expression of Ngef and Casp8ap2. Our results suggest that HOX protein-protein interactions without direct HOX DNA-binding may play a larger role in HOX transcriptional regulation than generally assumed, and DNA-binding appears critical for repression.
Collapse
Affiliation(s)
- Thomas M Williams
- Department of Human Genetics, University of Michigan, Ann Arbor, MI 48109-0618, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Hox genes encode transcription factors that control spatial patterning during embryogenesis. To date, downstream targets of Hox genes have proven difficult to identify. Here, we describe studies designed to identify target genes under the control of the murine transcription factor Hoxc8. We used a mouse 16,463 gene oligonucleotide microarray to identify mRNAs whose expression was altered by the overexpression of Hoxc8 in C57BL/6J mouse embryo fibroblasts (MEF) in cell culture (in vitro). We identified a total of 34 genes whose expression was changed by 2-fold or greater: 16 genes were up-regulated, and 18 genes were down-regulated. The majority of genes encoded proteins involved in critical biological processes, such as cell adhesion, migration, metabolism, apoptosis, and tumorigenesis. Two genes showed high levels of regulation: (i) secreted phosphoprotein 1 (Spp1), also known as osteopontin (OPN), was down-regulated 4.8-fold, and (ii) frizzled homolog 2 (Drosophila) (Fzd2) was up-regulated 4.4-fold. Chromatin immunoprecipitation (ChIP) analysis confirmed the direct interaction between the OPN promoter and Hoxc8 protein in vivo, supporting the view that OPN is a direct transcriptional target of Hoxc8.
Collapse
Affiliation(s)
- Haiyan Lei
- Department of Molecular, Cellular, and Developmental Biology, Yale University, 266 Whitney Avenue, New Haven, CT 06511, USA
| | | | | | | |
Collapse
|
50
|
Theoleyre S, Wittrant Y, Tat SK, Fortun Y, Redini F, Heymann D. The molecular triad OPG/RANK/RANKL: involvement in the orchestration of pathophysiological bone remodeling. Cytokine Growth Factor Rev 2004; 15:457-75. [PMID: 15561602 DOI: 10.1016/j.cytogfr.2004.06.004] [Citation(s) in RCA: 461] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The past decade has seen an explosion in the field of bone biology. The area of bone biology over this period of time has been marked by a number of key discoveries that have opened up entirely new areas for investigation. The recent identification of the receptor activator of nuclear factor kappaB ligand (RANKL), its cognate receptor RANK, and its decoy receptor osteoprotegerin (OPG) has led to a new molecular perspective on osteoclast biology and bone homeostasis. Specifically, the interaction between RANKL and RANK has been shown to be required for osteoclast differentiation. The third protagonist, OPG, acts as a soluble receptor antagonist for RANKL that prevents it from binding to and activating RANK. Any dysregulation of their respective expression leads to pathological conditions such as bone tumor-associated osteolysis, immune disease, or cardiovascular pathology. In this context, the OPG/RANK/RANKL triad opens novel therapeutic areas in diseases characterized by excessive bone resorption. The present article is an update and extension of an earlier review published by Kwan Tat et al. [Kwan Tat S, Padrines M, Theoleyre S, Heymann D, Fortun Y. IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev 2004;15:49-60].
Collapse
Affiliation(s)
- Sandrine Theoleyre
- EA 3822, INSERM ESPRI, Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Faculté de Médecine, 1 rue Gaston Veil, 44035 Nantes Cedex 1, France
| | | | | | | | | | | |
Collapse
|