1
|
Labelle-Dumais C, Mazur C, Kaya S, Obata Y, Lee B, Acevedo C, Alliston T, Gould DB. Skeletal pathology in mouse models of Gould syndrome is partially alleviated by genetically reducing TGFβ signaling. Matrix Biol 2024; 133:1-13. [PMID: 39097038 DOI: 10.1016/j.matbio.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/05/2024]
Abstract
Skeletal defects are hallmark features of many extracellular matrix (ECM) and collagen-related disorders. However, a biological function in bone has never been defined for the highly evolutionarily conserved type IV collagen. Collagen type IV alpha 1 (COL4A1) and alpha 2 (COL4A2) form α1α1α2 (IV) heterotrimers that represent a fundamental basement membrane constituent present in every organ of the body, including the skeleton. COL4A1 and COL4A2 mutations cause Gould syndrome, a variable and clinically heterogenous multisystem disorder generally characterized by the presence of cerebrovascular disease with ocular, renal, and muscular manifestations. We have previously identified elevated TGFβ signaling as a pathological insult resulting from Col4a1 mutations and demonstrated that reducing TGFβ signaling ameliorate ocular and cerebrovascular phenotypes in Col4a1 mutant mouse models of Gould syndrome. In this study, we describe the first characterization of skeletal defects in Col4a1 mutant mice that include a developmental delay in osteogenesis and structural, biomechanical and vascular alterations of mature bones. Using distinct mouse models, we show that allelic heterogeneity influences the presentation of skeletal pathology resulting from Col4a1 mutations. Importantly, we found that TGFβ target gene expression is elevated in developing bones from Col4a1 mutant mice and show that genetically reducing TGFβ signaling partially ameliorates skeletal manifestations. Collectively, these findings identify a novel and unsuspected role for type IV collagen in bone biology, expand the spectrum of manifestations associated with Gould syndrome to include skeletal abnormalities, and implicate elevated TGFβ signaling in skeletal pathogenesis in Col4a1 mutant mice.
Collapse
Affiliation(s)
- Cassandre Labelle-Dumais
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Courtney Mazur
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA
| | - Yoshihiro Obata
- Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Bryson Lee
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA
| | - Claire Acevedo
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; Materials Science Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Mechanical and Aerospace Engineering, University of California San Diego, San Diego, CA 92093, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California San Francisco, San Francisco, CA, 94143, USA; UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA 94143, USA
| | - Douglas B Gould
- Departments of Ophthalmology, University of California San Francisco, San Francisco, CA 94143, USA; Department of Anatomy, Institute for Human Genetics, Bakar Aging Research Institute, and Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
2
|
Goloe D, Gildor T, Ben-Tabou de-Leon S. Expression and Transcriptional Targets of TGFβ-RII in Paracentrotus lividus Larval Skeletogenesis. Genesis 2024; 62:e23614. [PMID: 39139086 DOI: 10.1002/dvg.23614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/23/2024] [Accepted: 07/25/2024] [Indexed: 08/15/2024]
Abstract
Organisms from the five kingdoms of life use minerals to harden their tissues and make teeth, shells and skeletons, in the process of biomineralization. The sea urchin larval skeleton is an excellent system to study the biological regulation of biomineralization and its evolution. The gene regulatory network (GRN) that controls sea urchin skeletogenesis is known in great details and shows similarity to the GRN that controls vertebrates' vascularization while it is quite distinct from the GRN that drives vertebrates' bone formation. Yet, transforming growth factor beta (TGF-β) signaling regulates both sea urchin and vertebrates' skeletogenesis. Here, we study the upstream regulation and identify transcriptional targets of TGF-β in the Mediterranean Sea urchin species, Paracentrotus lividus. TGF-βRII is transiently active in the skeletogenic cells downstream of vascular endothelial growth factor (VEGF) signaling, in P. lividus. Continuous perturbation of TGF-βRII activity significantly impairs skeletal elongation and the expression of key skeletogenic genes. Perturbation of TGF-βRII after skeletal initiation leads to a delay in skeletal elongation and minor changes in gene expression. TGF-β targets are distinct from its transcriptional targets during vertebrates' bone formation, suggesting that the role of TGF-β in biomineralization in these two phyla results from convergent evolution.
Collapse
Affiliation(s)
- Daniel Goloe
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, Israel
| | - Tsvia Gildor
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, Israel
| | - Smadar Ben-Tabou de-Leon
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Haifa, Israel
| |
Collapse
|
3
|
Dole NS, Betancourt-Torres A, Kaya S, Obata Y, Schurman CA, Yoon J, Yee CS, Khanal V, Luna CA, Carroll M, Salinas JJ, Miclau E, Acevedo C, Alliston T. High-fat and high-carbohydrate diets increase bone fragility through TGF-β-dependent control of osteocyte function. JCI Insight 2024; 9:e175103. [PMID: 39171528 PMCID: PMC11343608 DOI: 10.1172/jci.insight.175103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 07/03/2024] [Indexed: 08/23/2024] Open
Abstract
Obesity can increase the risk of bone fragility, even when bone mass is intact. This fragility stems from poor bone quality, potentially caused by deficiencies in bone matrix material properties. However, cellular and molecular mechanisms leading to obesity-related bone fragility are not fully understood. Using male mouse models of obesity, we discovered TGF-β signaling plays a critical role in mediating the effects of obesity on bone. High-carbohydrate and high-fat diets increase TGF-β signaling in osteocytes, which impairs their mitochondrial function, increases cellular senescence, and compromises perilacunar/canalicular remodeling and bone quality. By specifically inhibiting TGF-β signaling in mouse osteocytes, some of the negative effects of high-fat and high-carbohydrate diets on bones, including the lacunocanalicular network, perilacunar/canalicular remodeling, senescence, and mechanical properties such as yield stress, were mitigated. DMP1-Cre-mediated deletion of TGF-β receptor II also blunted adverse effects of high-fat and high-carbohydrate diets on energy balance and metabolism. These findings suggest osteocytes are key in controlling bone quality in response to high-fat and high-carbohydrate diets. Calibrating osteocyte function could mitigate bone fragility associated with metabolic diseases while reestablishing energy balance.
Collapse
Affiliation(s)
- Neha S. Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock Arkansas, USA
| | - Andrés Betancourt-Torres
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Yoshihiro Obata
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| | - Jihee Yoon
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Cristal S. Yee
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Vivek Khanal
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock Arkansas, USA
| | - Clarissa Aguirre Luna
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Madeline Carroll
- Department of Physiology and Cell Biology, University of Arkansas for Medical Sciences, Little Rock Arkansas, USA
| | - Jennifer J. Salinas
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Elizabeth Miclau
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Claire Acevedo
- Department of Mechanical Engineering, University of Utah, Salt Lake City, Utah, USA
- Department of Biomedical Engineering, University of Utah, Salt Lake City, Utah, USA
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, San Diego, California, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, San Francisco, California, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, California, USA
| |
Collapse
|
4
|
Kaspiris A, Vasiliadis ES, Tsalimas G, Melissaridou D, Lianou I, Panagopoulos F, Katzouraki G, Vavourakis M, Kolovos I, Savvidou OD, Papadimitriou E, Pneumaticos SG. Unraveling the Link of Altered TGFβ Signaling with Scoliotic Vertebral Malformations in Osteogenesis Imperfecta: A Comprehensive Review. J Clin Med 2024; 13:3484. [PMID: 38930011 PMCID: PMC11204596 DOI: 10.3390/jcm13123484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 05/27/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
Osteogenesis Imperfecta (OI) is a genetic disorder caused by mutations in genes responsible for collagen synthesis or polypeptides involved in the formation of collagen fibers. Its predominant skeletal complication is scoliosis, impacting 25 to 80% of OI patients. Vertebral deformities of the scoliotic curves in OI include a variety of malformations such as codfish, wedged-shaped vertebrae or platyspondyly, craniocervical junction abnormalities, and lumbosacral spondylolysis and spondylolisthesis. Although the precise pathophysiology of these spinal deformities remains unclear, anomalies in bone metabolism have been implicated in the progression of scoliotic curves. Bone Mineral Density (BMD) measurements have demonstrated a significant reduction in the Z-score, indicating osteoporosis and a correlation with the advancement of scoliosis. Factors such as increased mechanical strains, joint hypermobility, lower leg length discrepancy, pelvic obliquity, spinal ligament hypermobility, or vertebrae microfractures may also contribute to the severity of scoliosis. Histological vertebral analysis has confirmed that changes in trabecular microarchitecture, associated with inadequate bone turnover, indicate generalized bone metabolic defects in OI. At the molecular level, the upregulation of Transforming Growth factor-β (TGFβ) signaling in OI can lead to disturbed bone turnover and changes in muscle mass and strength. Understanding the relationship between spinal clinical features and molecular pathways could unveil TGFβ -related molecular targets, paving the way for novel therapeutic approaches in OI.
Collapse
Affiliation(s)
- Angelos Kaspiris
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
- Laboratory of Molecular Pharmacology, Group for Orthopaedic Research, School of Health Sciences, University of Patras, 26504 Patras, Greece;
| | - Elias S. Vasiliadis
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
| | - Georgios Tsalimas
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
| | - Dimitra Melissaridou
- First Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “ATTIKON” University Hospital, Rimini 1, 12462 Athens, Greece; (D.M.); (O.D.S.)
| | - Ioanna Lianou
- Department of Orthopaedic Surgery, “Rion” University Hospital and Medical School, School of Health Sciences, University of Patras, 26504 Patras, Greece; (I.L.); (F.P.)
| | - Fotios Panagopoulos
- Department of Orthopaedic Surgery, “Rion” University Hospital and Medical School, School of Health Sciences, University of Patras, 26504 Patras, Greece; (I.L.); (F.P.)
| | - Galateia Katzouraki
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
| | - Michail Vavourakis
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
| | - Ioannis Kolovos
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
| | - Olga D. Savvidou
- First Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “ATTIKON” University Hospital, Rimini 1, 12462 Athens, Greece; (D.M.); (O.D.S.)
| | - Evangelia Papadimitriou
- Laboratory of Molecular Pharmacology, Group for Orthopaedic Research, School of Health Sciences, University of Patras, 26504 Patras, Greece;
| | - Spiros G. Pneumaticos
- Third Department of Orthopaedic Surgery, School of Medicine, National and Kapodistrian University of Athens, “KAT” General Hospital, Nikis 2, 14561 Athens, Greece; (E.S.V.); (G.T.); (G.K.); (M.V.); (I.K.); (S.G.P.)
| |
Collapse
|
5
|
Myint O, Sakunrangsit N, Pholtaisong J, Toejing P, Pho-on P, Leelahavanichkul A, Sridurongrit S, Aporntewan C, Greenblatt MB, Lotinun S. Differential Gene Expression Involved in Bone Turnover of Mice Expressing Constitutively Active TGFβ Receptor Type I. Int J Mol Sci 2024; 25:5829. [PMID: 38892016 PMCID: PMC11173332 DOI: 10.3390/ijms25115829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Transforming growth factor beta (TGF-β) is ubiquitously found in bone and plays a key role in bone turnover. Mice expressing constitutively active TGF-β receptor type I (Mx1;TβRICA mice) are osteopenic. Here, we identified the candidate genes involved in bone turnover in Mx1;TβRICA mice using RNA sequencing analysis. A total of 285 genes, including 87 upregulated and 198 downregulated genes, were differentially expressed. According to the KEGG analysis, some genes were involved in osteoclast differentiation (Fcgr4, Lilrb4a), B cell receptor signaling (Cd72, Lilrb4a), and neutrophil extracellular trap formation (Hdac7, Padi4). Lilrb4 is related to osteoclast inhibition protein, whereas Hdac7 is a Runx2 corepressor that regulates osteoblast differentiation. Silencing Lilrb4 increased the number of osteoclasts and osteoclast marker genes. The knocking down of Hdac7 increased alkaline phosphatase activity, mineralization, and osteoblast marker genes. Therefore, our present study may provide an innovative idea for potential therapeutic targets and pathways in TβRI-associated bone loss.
Collapse
Affiliation(s)
- Ohnmar Myint
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (O.M.); (N.S.); (J.P.); (P.T.); (P.P.-o.)
| | - Nithidol Sakunrangsit
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (O.M.); (N.S.); (J.P.); (P.T.); (P.P.-o.)
| | - Jatuphol Pholtaisong
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (O.M.); (N.S.); (J.P.); (P.T.); (P.P.-o.)
| | - Parichart Toejing
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (O.M.); (N.S.); (J.P.); (P.T.); (P.P.-o.)
| | - Pinyada Pho-on
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (O.M.); (N.S.); (J.P.); (P.T.); (P.P.-o.)
| | - Asada Leelahavanichkul
- Division of Immunology, Department of Microbiology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Somyoth Sridurongrit
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand;
| | - Chatchawit Aporntewan
- Department of Mathematics and Computer Science, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Matthew B. Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY 10065, USA;
- Research Division, Hospital for Special Surgery, New York, NY 10065, USA
| | - Sutada Lotinun
- Center of Excellence in Skeletal Disorders and Enzyme Reaction Mechanism, Department of Physiology, Faculty of Dentistry, Chulalongkorn University, Bangkok 10330, Thailand; (O.M.); (N.S.); (J.P.); (P.T.); (P.P.-o.)
| |
Collapse
|
6
|
Schurman CA, Kaya S, Dole N, Luna NMM, Castillo N, Potter R, Rose JP, Bons J, King CD, Burton JB, Schilling B, Melov S, Tang S, Schaible E, Alliston T. Aging impairs the osteocytic regulation of collagen integrity and bone quality. Bone Res 2024; 12:13. [PMID: 38409111 PMCID: PMC10897167 DOI: 10.1038/s41413-023-00303-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 10/31/2023] [Accepted: 11/13/2023] [Indexed: 02/28/2024] Open
Abstract
Poor bone quality is a major factor in skeletal fragility in elderly individuals. The molecular mechanisms that establish and maintain bone quality, independent of bone mass, are unknown but are thought to be primarily determined by osteocytes. We hypothesize that the age-related decline in bone quality results from the suppression of osteocyte perilacunar/canalicular remodeling (PLR), which maintains bone material properties. We examined bones from young and aged mice with osteocyte-intrinsic repression of TGFβ signaling (TβRIIocy-/-) that suppresses PLR. The control aged bone displayed decreased TGFβ signaling and PLR, but aging did not worsen the existing PLR suppression in male TβRIIocy-/- bone. This relationship impacted the behavior of collagen material at the nanoscale and tissue scale in macromechanical tests. The effects of age on bone mass, density, and mineral material behavior were independent of osteocytic TGFβ. We determined that the decline in bone quality with age arises from the loss of osteocyte function and the loss of TGFβ-dependent maintenance of collagen integrity.
Collapse
Affiliation(s)
- Charles A Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, 94143, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, 94143, USA
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, 94143, USA
| | - Neha Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, 94143, USA
| | - Nadja M Maldonado Luna
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, 94143, USA
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, 94143, USA
| | - Natalia Castillo
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, 94143, USA
| | - Ryan Potter
- Washington University in St Louis, Department of Orthopedics, St. Louis, MO, 63130, USA
| | - Jacob P Rose
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Joanna Bons
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Jordan B Burton
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | | | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA, 94945, USA
| | - Simon Tang
- Washington University in St Louis, Department of Orthopedics, St. Louis, MO, 63130, USA
| | - Eric Schaible
- Advanced Light Source, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA, 94143, USA.
- UC Berkeley/UCSF Graduate Program in Bioengineering, San Francisco, CA, 94143, USA.
| |
Collapse
|
7
|
Ge R, Huang GM. Targeting transforming growth factor beta signaling in metastatic osteosarcoma. J Bone Oncol 2023; 43:100513. [PMID: 38021074 PMCID: PMC10666000 DOI: 10.1016/j.jbo.2023.100513] [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: 10/03/2023] [Revised: 10/28/2023] [Accepted: 11/07/2023] [Indexed: 12/01/2023] Open
Abstract
Osteosarcoma is a rare type of bone cancer, and half of the cases affect children and adolescents younger than 20 years of age. Despite intensive efforts to improve both chemotherapeutics and surgical management, the clinical outcome for metastatic osteosarcoma remains poor. Transforming growth factor β (TGF-β) is one of the most abundant growth factors in bones. The TGF-β signaling pathway has complex and contradictory roles in the pathogenesis of human cancers. TGF-β is primarily a tumor suppressor that inhibits proliferation and induces apoptosis of premalignant epithelial cells. In the later stages of cancer progression, however, TGF-β functions as a metastasis promoter by promoting tumor growth, inducing epithelial-mesenchymal transition (EMT), blocking antitumor immune responses, increasing tumor-associated fibrosis, and enhancing angiogenesis. In contrast with the dual effects of TGF-β on carcinoma (epithelial origin) progression, TGF-β seems to mainly have a pro-tumoral effect on sarcomas including osteosarcoma (mesenchymal origin). Many drugs that target TGF-β signaling have been developed: neutralizing antibodies that prevent TGF-β binding to receptor complexes; ligand trap employing recombinant Fc-fusion proteins containing the soluble ectodomain of either type II (TβRII) or the type III receptor ((TβRIII), preventing TGF-β from binding to its receptors; antisense nucleotides that reduce TGF-β expression at the transcriptional/translational level; small molecule inhibitors of serine/threonine kinases of the type I receptor (TβRI) preventing downstream signaling; and vaccines that contain cell lines transfected with TβRII antisense genes, or target furin convertase, resulting in reduced TGF-β signaling. TGF-β antagonists have been shown to have effects on osteosarcoma in vitro and in vivo. One of the small molecule TβRI inhibitors, Vactosertib, is currently undergoing a phase 1/2 clinical trial to evaluate its effect on osteosarcoma. Several phase 1/2/3 clinical trials have shown TGF-β antagonists are safe and well tolerated. For instance, Luspatercept, a TGF-β ligand trap, has been approved by the FDA for the treatment of anemia associated with myeloid dysplastic syndrome (MDS) with ring sideroblasts/mutated SF3B1 with acceptable safety. Clinical trials evaluating the long-term safety of Luspatercept are in process.
Collapse
Affiliation(s)
- Rongrong Ge
- Hillman Cancer Center at Central Pennsylvania, University of Pittsburg Medical Center, Harrisburg, PA, 17109, USA
| | - Gavin M. Huang
- Harrisburg Academy School, 10 Erford Rd, Wormleysburg, PA, 17043, USA
| |
Collapse
|
8
|
Asparuhova MB, Riedwyl D, Aizawa R, Raabe C, Couso-Queiruga E, Chappuis V. Local Concentrations of TGF-β1 and IGF-1 Appear Determinant in Regulating Bone Regeneration in Human Postextraction Tooth Sockets. Int J Mol Sci 2023; 24:ijms24098239. [PMID: 37175951 PMCID: PMC10179638 DOI: 10.3390/ijms24098239] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/28/2023] [Accepted: 04/29/2023] [Indexed: 05/15/2023] Open
Abstract
Healing after tooth extraction involves a series of reparative processes affecting both alveolar bone and soft tissues. The aim of the present study was to investigate whether activation of molecular signals during the healing process confers a regenerative advantage to the extraction socket soft tissue (ESsT) at 8 weeks of healing. Compared to subepithelial connective tissue graft (CTG), qRT-PCR analyses revealed a dramatic enrichment of the ESsT in osteogenic differentiation markers. However, ESsT and CTG shared characteristics of nonspecialized soft connective tissue by expressing comparable levels of genes encoding abundant extracellular matrix (ECM) proteins. Genes encoding the transforming growth factor-β1 (TGF-β1) and its receptors were strongly enriched in the CTG, whereas the transcript for the insulin-like growth factor-1 (IGF-1) showed significantly high and comparable expression in both tissues. Mechanical stimulation, by the means of cyclic strain or matrix stiffness applied to primary ESsT cells (ESsT-C) and CTG fibroblasts (CTG-F) extracted from the tissue samples, revealed that stress-induced TGF-β1 not exceeding 2.3 ng/mL, as measured by ELISA, in combination with IGF-1 up to 2.5 ng/mL was able to induce the osteogenic potential of ESsT-Cs. However, stiff matrices (50 kPa), upregulating the TGF-β1 expression up to 6.6 ng/mL, caused downregulation of osteogenic gene expression in the ESsT-Cs. In CTG-Fs, endogenous or stress-induced TGF-β1 ≥ 4.6 ng/mL was likely responsible for the complete lack of osteogenesis. Treatment of ESsT-Cs with TGF-β1 and IGF-1 proved that, at specific concentrations, the two growth factors exhibited either an inductive-synergistic or a suppressive activity, thus determining the osteogenic and mineralization potential of ESsT-Cs. Taken together, our data strongly warrant the clinical exploration of ESsT as a graft in augmentative procedures during dental implant placement surgeries.
Collapse
Affiliation(s)
- Maria B Asparuhova
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Freiburgstrasse 3, 3010 Bern, Switzerland
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
| | - Dominic Riedwyl
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Freiburgstrasse 3, 3010 Bern, Switzerland
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
| | - Ryo Aizawa
- Laboratory of Oral Cell Biology, Dental Research Center, School of Dental Medicine, University of Bern, Freiburgstrasse 3, 3010 Bern, Switzerland
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
- Department of Periodontology, School of Dentistry, Showa University, 2-1-1 Kitasenzoku, Ohta-ku, Tokyo 145-8515, Japan
| | - Clemens Raabe
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
| | - Emilio Couso-Queiruga
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
| | - Vivianne Chappuis
- Department of Oral Surgery and Stomatology, School of Dental Medicine, University of Bern, Freiburgstrasse 7, 3010 Bern, Switzerland
| |
Collapse
|
9
|
Yalcin-Ülker GM, Günbatan M, Duygu G, Soluk-Tekkesin M, Özcakir-Tomruk C. Could Local Application of Hypoxia Inducible Factor 1-α Enhancer Deferoxamine Be Promising for Preventing of Medication-Related Osteonecrosis of the Jaw? Biomedicines 2023; 11:biomedicines11030758. [PMID: 36979736 PMCID: PMC10045901 DOI: 10.3390/biomedicines11030758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/25/2023] [Accepted: 02/26/2023] [Indexed: 03/06/2023] Open
Abstract
This experimental study investigates the prophylactic effect of deferoxamine (DFO) on medication-related osteonecrosis of the jaw (MRONJ). Thirty-six female Sprague Dawley rats received zoledronic acid (ZA) for eight weeks to create an osteonecrosis model. DFO was locally applied into the extraction sockets with gelatin sponge (GS) carriers to prevent MRONJ. The specimens were histopathologically and histomorphometrically evaluated. Hypoxia-inducible factor 1-alpha (HIF-1α) protein levels in the extraction sockets were quantified. New bone formation rate differed significantly between groups (p = 0.005). Newly formed bone ratios in the extraction sockets did not differ significantly between the control group and the GS (p = 1), GS/DFO (p = 0.749), ZA (p = 0.105), ZA-GS (p = 0.474), and ZA-GS/DFO (p = 1) groups. While newly formed bone rates were higher in the ZA-GS and ZA-GS/DFO groups than in the ZA group, the differences were not significant. HIF-1α levels differed significantly between groups (p < 0.001) and were significantly higher in the DFO and ZA-GS/DFO groups than in the control group (p = 0.001 and p = 0.004, respectively). While HIF-1α levels were higher in the ZA-GS/DFO group than in the ZA group, the difference was not significant. While HIF-1α protein levels and new bone formation rate were elevated in the DFO-treated group, the effect was not significant. Further large-scale studies are needed to understand DFO’s preventative effects on MRONJ and the role of HIF-1α in MRONJ pathogenesis.
Collapse
Affiliation(s)
- Gül Merve Yalcin-Ülker
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Istanbul Okan University, Istanbul 34947, Türkiye
- Correspondence: or
| | - Murat Günbatan
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Istanbul Okan University, Istanbul 34947, Türkiye
| | - Gonca Duygu
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Tekirdag Namık Kemal University, Tekirdag 59030, Türkiye
| | - Merva Soluk-Tekkesin
- Department of Tumour Pathology, Institute of Oncology, Istanbul University, Istanbul 34093, Türkiye
| | - Ceyda Özcakir-Tomruk
- Oral and Maxillofacial Surgery Department, Faculty of Dentistry, Yeditepe University, Istanbul 34728, Türkiye
| |
Collapse
|
10
|
Chondrocyte Hypertrophy in Osteoarthritis: Mechanistic Studies and Models for the Identification of New Therapeutic Strategies. Cells 2022; 11:cells11244034. [PMID: 36552796 PMCID: PMC9777397 DOI: 10.3390/cells11244034] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 12/08/2022] [Indexed: 12/16/2022] Open
Abstract
Articular cartilage shows limited self-healing ability owing to its low cellularity and avascularity. Untreated cartilage defects display an increased propensity to degenerate, leading to osteoarthritis (OA). During OA progression, articular chondrocytes are subjected to significant alterations in gene expression and phenotype, including a shift towards a hypertrophic-like state (with the expression of collagen type X, matrix metalloproteinases-13, and alkaline phosphatase) analogous to what eventuates during endochondral ossification. Present OA management strategies focus, however, exclusively on cartilage inflammation and degradation. A better understanding of the hypertrophic chondrocyte phenotype in OA might give new insights into its pathogenesis, suggesting potential disease-modifying therapeutic approaches. Recent developments in the field of cellular/molecular biology and tissue engineering proceeded in the direction of contrasting the onset of this hypertrophic phenotype, but knowledge gaps in the cause-effect of these processes are still present. In this review we will highlight the possible advantages and drawbacks of using this approach as a therapeutic strategy while focusing on the experimental models necessary for a better understanding of the phenomenon. Specifically, we will discuss in brief the cellular signaling pathways associated with the onset of a hypertrophic phenotype in chondrocytes during the progression of OA and will analyze in depth the advantages and disadvantages of various models that have been used to mimic it. Afterwards, we will present the strategies developed and proposed to impede chondrocyte hypertrophy and cartilage matrix mineralization/calcification. Finally, we will examine the future perspectives of OA therapeutic strategies.
Collapse
|
11
|
Mavroudis PD, Pillai N, Wang Q, Pouzin C, Greene B, Fretland J. A multi-model approach to predict efficacious clinical dose for an anti-TGF-β antibody (GC2008) in the treatment of osteogenesis imperfecta. CPT Pharmacometrics Syst Pharmacol 2022; 11:1485-1496. [PMID: 36004727 PMCID: PMC9662198 DOI: 10.1002/psp4.12857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 08/02/2022] [Accepted: 08/11/2022] [Indexed: 11/29/2022] Open
Abstract
Osteogenesis imperfecta (OI) is a heterogeneous group of inherited bone dysplasias characterized by reduced skeletal mass and bone fragility. Although the primary manifestation of the disease involves the skeleton, OI is a generalized connective tissue disorder that requires a multidisciplinary treatment approach. Recent studies indicate that application of a transforming growth factor beta (TGF-β) neutralizing antibody increased bone volume fraction (BVF) and strength in an OI mouse model and improved bone mineral density (BMD) in a small cohort of patients with OI. In this work, we have developed a multitiered quantitative pharmacology approach to predict human efficacious dose of a new anti-TGF-β antibody drug candidate (GC2008). This method aims to translate GC2008 pharmacokinetic/pharmacodynamic (PK/PD) relationship in patients, using a number of appropriate mathematical models and available preclinical and clinical data. Compartmental PK linked with an indirect PD effect model was used to characterize both pre-clinical and clinical PK/PD data and predict a GC2008 dose that would significantly increase BMD or BVF in patients with OI. Furthermore, a physiologically-based pharmacokinetic model incorporating GC2008 and the body's physiological properties was developed and used to predict a GC2008 dose that would decrease the TGF-β level in bone to that of healthy individuals. By using multiple models, we aim to reveal information for different aspects of OI disease that will ultimately lead to a more informed dose projection of GC2008 in humans. The different modeling efforts predicted a similar range of pharmacologically relevant doses in patients with OI providing an informed approach for an early clinical dose setting.
Collapse
Affiliation(s)
| | - Nikhil Pillai
- Quantitative PharmacologyDMPK, Sanofi USWalthamMassachusettsUSA
| | | | | | - Benjamin Greene
- Rare and Neurologic Diseases ResearchSanofiFraminghamMassachusettsUSA
| | | |
Collapse
|
12
|
Atomic force microscopy (AFM) and its applications to bone-related research. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2022; 176:52-66. [DOI: 10.1016/j.pbiomolbio.2022.10.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 09/19/2022] [Accepted: 10/07/2022] [Indexed: 11/06/2022]
|
13
|
Kaya S, Schurman CA, Dole NS, Evans DS, Alliston T. Prioritization of Genes Relevant to Bone Fragility Through the Unbiased Integration of Aging Mouse Bone Transcriptomics and Human GWAS Analyses. J Bone Miner Res 2022; 37:804-817. [PMID: 35094432 PMCID: PMC9018503 DOI: 10.1002/jbmr.4516] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 12/20/2021] [Accepted: 01/17/2022] [Indexed: 11/10/2022]
Abstract
Identifying new genetic determinants of bone mineral density (BMD) and fracture promises to yield improved diagnostics and therapies for bone fragility. However, prioritizing candidate genes from genome-wide screens can be challenging. To overcome this challenge, we prioritized mouse genes that are differentially expressed in aging mouse bone based on whether their human homolog is associated with human BMD and/or fracture. Unbiased RNA-seq analysis of young and old male C57BL/6 mouse cortical bone identified 1499, 1685, and 5525 differentially expressed genes (DEGs) in 1, 2, and 2.5-year-old bone, relative to 2-month-old bone, respectively. Gene-based scores for heel ultrasound bone mineral density (eBMD) and fracture were estimated using published genome-wide association studies (GWAS) results of these traits in the UK Biobank. Enrichment analysis showed that mouse bone DEG sets for all three age groups, relative to young bone, are significantly enriched for eBMD, but only the oldest two DEG sets are enriched for fracture. Using gene-based scores, this approach prioritizes among thousands of DEGs by a factor of 5- to 100-fold, yielding 10 and 21 genes significantly associated with fracture in the two oldest groups of mouse DEGs. Though these genes were not the most differentially expressed, they included Sost, Lrp5, and others with well-established functions in bone. Several others have, as yet, unknown roles in the skeleton. Therefore, this study accelerates identification of new genetic determinants of bone fragility by prioritizing a clinically relevant and experimentally tractable number of candidate genes for functional analysis. Finally, we provide a website (www.mouse2human.org) to enable other researchers to easily apply our strategy. © 2022 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Serra Kaya
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Charles A. Schurman
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA
| | - Neha S. Dole
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute, San Francisco, CA
| | - Tamara Alliston
- Department of Orthopaedic Surgery, University of California, San Francisco, CA
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA
| |
Collapse
|
14
|
Guerrerio AL, Mateja A, Rasooly M, Levin S, Magnani A, Dempsey C, MacCarrick G, Dietz HC, Brittain E, Boyce AM, Frischmeyer-Guerrerio PA. Predictors of low bone density and fracture risk in Loeys-Dietz syndrome. Genet Med 2022; 24:419-429. [PMID: 34906513 PMCID: PMC11009834 DOI: 10.1016/j.gim.2021.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/26/2021] [Accepted: 10/04/2021] [Indexed: 11/17/2022] Open
Abstract
PURPOSE Loeys-Dietz syndrome (LDS) is a connective tissue disorder affecting multiple organ systems, including bone. METHODS We defined the bone phenotype and clinical predictors of low bone density and fracture risk in 77 patients with LDS type 1 to type 5. RESULTS Patients with LDS had dual-energy x-ray absorptiometry (DXA) Z-scores significantly < 0, and 50% of children and 9% of adults had Z-scores < -2. Sixty percent of patients had ≥1 fracture, and 24% of patients with spinal x-rays scans showed spinal compression fractures. Lower body mass index, asthma, male sex and eosinophilic gastrointestinal disease were correlated with lower DXA Z-scores. The count of 5 LDS-associated skeletal features (scoliosis, pes planus, arachnodactyly, spondylolisthesis, and camptodactyly) in patients with LDS was correlated with DXA Z-score. Adults with ≥1 skeletal features had DXA Z-scores significantly < 0, and children with >2 features had DXA Z-score significantly < -2. Bone turnover markers suggest accelerated bone resorption. Data from 5 patients treated with bisphosphonates suggest a beneficial effect. CONCLUSION All LDS types are associated with reduced bone density and increased risk of fracture, which may be due to increased bone resorption. Clinical features can predict a subgroup of patients at highest risk of low bone density and fracture risk.
Collapse
Affiliation(s)
- Anthony L Guerrerio
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Allyson Mateja
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory, Leidos Biomedical Research, Inc, Frederick, MD
| | - Marjohn Rasooly
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Samara Levin
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Alaina Magnani
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Caeden Dempsey
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Gretchen MacCarrick
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Harry C Dietz
- McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD; Howard Hughes Medical Institute, Chevy Chase, MD
| | - Erica Brittain
- Biostatistics Research Branch (BRB), National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Alison M Boyce
- Metabolic Bone Disorders Unit, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD.
| | - Pamela A Frischmeyer-Guerrerio
- The Laboratory of Allergic Diseases, National Institutes of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
15
|
Trivedi T, Guise TA. Systemic effects of abnormal bone resorption on muscle, metabolism, and cognition. Bone 2022; 154:116245. [PMID: 34718221 DOI: 10.1016/j.bone.2021.116245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 10/21/2021] [Accepted: 10/25/2021] [Indexed: 12/11/2022]
Abstract
Skeletal tissue is dynamic, undergoing constant remodeling to maintain musculoskeletal integrity and balance in the human body. Recent evidence shows that apart from maintaining homeostasis in the local microenvironment, the skeleton systemically affects other tissues. Several cancer-associated and noncancer-associated bone disorders can disrupt the physiological homeostasis locally in the bone microenvironment and indirectly contribute to dysregulation of systemic body function. The systemic effects of bone on the regulation of distant organ function have not been widely explored. Recent evidence suggests that bone can interact with skeletal muscle, pancreas, and brain by releasing factors from mineralized bone matrix. Currently available bone-targeting therapies such as bisphosphonates and denosumab inhibit bone resorption, decrease morbidity associated with bone destruction, and improve survival. Bisphosphonates have been a standard treatment for bone metastases, osteoporosis, and cancer treatment-induced bone diseases. The extraskeletal effects of bisphosphonates on inhibition of tumor growth are known. However, our knowledge of the effects of bisphosphonates on muscle weakness, hyperglycemia, and cognitive defects is currently evolving. To be able to identify the molecular link between bone and distant organs during abnormal bone resorption and then treat these abnormalities and prevent their systemic effects could improve survival benefits. The current review highlights the link between bone resorption and its systemic effects on muscle, pancreas, and brain.
Collapse
Affiliation(s)
- Trupti Trivedi
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| | - Theresa A Guise
- Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, United States of America.
| |
Collapse
|
16
|
Sahbani K, Cardozo CP, Bauman WA, Tawfeek HA. Inhibition of TGF-β Signaling Attenuates Disuse-induced Trabecular Bone Loss After Spinal Cord Injury in Male Mice. Endocrinology 2022; 163:bqab230. [PMID: 34791098 DOI: 10.1210/endocr/bqab230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Indexed: 11/19/2022]
Abstract
Bone loss is one of the most common complications of immobilization after spinal cord injury (SCI). Whether transforming growth factor (TGF)-β signaling plays a role in SCI-induced disuse bone loss has not been determined. Thus, 16-week-old male mice underwent sham or spinal cord contusion injury to cause complete hindlimb paralysis. Five days later, 10 mg/kg/day control (IgG) or anti-TGF-β1,2,3 neutralizing antibody (1D11) was administered twice weekly for 4 weeks. Femurs were examined by micro-computed tomography (micro-CT) scanning and histology. Bone marrow (BM) supernatants were analyzed by enzyme-linked immunosorbent assay for levels of procollagen type 1 intact N-terminal propeptide (P1NP), tartrate-resistant acid phosphatase (TRAcP-5b), receptor activator of nuclear factor-kappa B ligand (RANKL), osteoprotegerin (OPG), and prostaglandin E2 (PGE2). Distal femoral micro-CT analysis showed that SCI-1D11 mice had significantly (P < .05) attenuated loss of trabecular fractional bone volume (123% SCI-1D11 vs 69% SCI-IgG), thickness (98% vs 81%), and connectivity (112% vs 69%) and improved the structure model index (2.1 vs 2.7). Histomorphometry analysis revealed that osteoclast numbers were lower in the SCI-IgG mice than in sham-IgG control. Biochemically, SCI-IgG mice had higher levels of P1NP and PGE2 but similar TRAcP-5b and RANKL/OPG ratio to the sham-IgG group. The SCI-1D11 group exhibited higher levels of P1NP but similar TRAcP-5b, RANKL/OPG ratio, and PGE2 to the sham-1D11 group. Furthermore, 1D11 treatment prevented SCI-induced hyperphosphorylation of tau protein in osteocytes, an event that destabilizes the cytoskeleton. Together, inhibition of TGF-β signaling after SCI protects trabecular bone integrity, likely by balancing bone remodeling, inhibiting PGE2 elevation, and preserving the osteocyte cytoskeleton.
Collapse
Affiliation(s)
- Karim Sahbani
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
| | - Christopher P Cardozo
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Institute for Systems Biomedicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - William A Bauman
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Department of Rehabilitation Medicine and Human Performance, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mount Sinai Institute for Systems Biomedicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Hesham A Tawfeek
- National Center for the Medical Consequences of Spinal Cord Injury, James J Peters Veterans Affairs Medical Center, Bronx, NY 10468, USA
- Bronx Veterans Medical Research Foundation, Bronx, NY 10468, USA
- Department of Medicine, The Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
17
|
Lehmann TP, Guderska U, Kałek K, Marzec M, Urbanek A, Czernikiewicz A, Sąsiadek M, Karpiński P, Pławski A, Głowacki M, Jagodziński PP. The Regulation of Collagen Processing by miRNAs in Disease and Possible Implications for Bone Turnover. Int J Mol Sci 2021; 23:91. [PMID: 35008515 PMCID: PMC8745169 DOI: 10.3390/ijms23010091] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022] Open
Abstract
This article describes several recent examples of miRNA governing the regulation of the gene expression involved in bone matrix construction. We present the impact of miRNA on the subsequent steps in the formation of collagen type I. Collagen type I is a main factor of mechanical bone stiffness because it constitutes 90-95% of the organic components of the bone. Therefore, the precise epigenetic regulation of collagen formation may have a significant influence on bone structure. We also describe miRNA involvement in the expression of genes, the protein products of which participate in collagen maturation in various tissues and cancer cells. We show how non-collagenous proteins in the extracellular matrix are epigenetically regulated by miRNA in bone and other tissues. We also delineate collagen mineralisation in bones by factors that depend on miRNA molecules. This review reveals the tissue variability of miRNA regulation at different levels of collagen maturation and mineralisation. The functionality of collagen mRNA regulation by miRNA, as proven in other tissues, has not yet been shown in osteoblasts. Several collagen-regulating miRNAs are co-expressed with collagen in bone. We suggest that collagen mRNA regulation by miRNA could also be potentially important in bone metabolism.
Collapse
Affiliation(s)
- Tomasz P. Lehmann
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| | - Urszula Guderska
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| | - Klaudia Kałek
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| | - Maria Marzec
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| | - Agnieszka Urbanek
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| | - Alicja Czernikiewicz
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| | - Maria Sąsiadek
- Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.S.); (P.K.)
| | - Paweł Karpiński
- Department of Genetics, Wroclaw Medical University, 50-368 Wroclaw, Poland; (M.S.); (P.K.)
| | - Andrzej Pławski
- Institute of Human Genetics, Polish Academy of Sciences, 60-479 Poznan, Poland;
| | - Maciej Głowacki
- Department of Paediatric Orthopaedics and Traumatology, Poznan University of Medical Sciences, 61-545 Poznan, Poland;
| | - Paweł P. Jagodziński
- Department of Biochemistry and Molecular Biology, Poznan University of Medical Sciences, 60-781 Poznan, Poland; (U.G.); (K.K.); (M.M.); (A.U.); (A.C.); (P.P.J.)
| |
Collapse
|
18
|
Carter A, Popowski K, Cheng K, Greenbaum A, Ligler FS, Moatti A. Enhancement of Bone Regeneration Through the Converse Piezoelectric Effect, A Novel Approach for Applying Mechanical Stimulation. Bioelectricity 2021; 3:255-271. [PMID: 35018335 PMCID: PMC8742263 DOI: 10.1089/bioe.2021.0019] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Serious bone injuries have devastating effects on the lives of patients including limiting working ability and high cost. Orthopedic implants can aid in healing injuries to an extent that exceeds the natural regenerative capabilities of bone to repair fractures or large bone defects. Autografts and allografts are the standard implants used, but disadvantages such as donor site complications, a limited quantity of transplantable bone, and high costs have led to an increased demand for synthetic bone graft substitutes. However, replicating the complex physiological properties of biological bone, much less recapitulating its complex tissue functions, is challenging. Extensive efforts to design biocompatible implants that mimic the natural healing processes in bone have led to the investigation of piezoelectric smart materials because the bone has natural piezoelectric properties. Piezoelectric materials facilitate bone regeneration either by accumulating electric charge in response to mechanical stress, which mimics bioelectric signals through the direct piezoelectric effect or by providing mechanical stimulation in response to electrical stimulation through the converse piezoelectric effect. Although both effects are beneficial, the converse piezoelectric effect can address bone atrophy from stress shielding and immobility by improving the mechanical response of a healing defect. Mechanical stimulation has a positive impact on bone regeneration by activating cellular pathways that increase bone formation and decrease bone resorption. This review will highlight the potential of the converse piezoelectric effect to enhance bone regeneration by discussing the activation of beneficial cellular pathways, the properties of piezoelectric biomaterials, and the potential for the more effective administration of the converse piezoelectric effect using wireless control.
Collapse
Affiliation(s)
- Amber Carter
- Department of Biological Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
| | - Kristen Popowski
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
| | - Ke Cheng
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, North Carolina, USA
- Division of Pharmacoengineering and Molecular Pharmaceutics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Alon Greenbaum
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Frances S. Ligler
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| | - Adele Moatti
- Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina, USA
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill and North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
19
|
Muñoz A, Docaj A, Ugarteburu M, Carriero A. Poor bone matrix quality: What can be done about it? Curr Osteoporos Rep 2021; 19:510-531. [PMID: 34414561 DOI: 10.1007/s11914-021-00696-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF THE REVIEW Bone's ability to withstand load resisting fracture and adapting to it highly depends on the quality of its matrix and its regulators. This review focuses on the contribution of bone quality to fracture resistance and possible therapeutic targets for skeletal fragility in aging and disease. RECENT FINDINGS The highly organized, hierarchical composite structure of bone extracellular matrix together with its (re)modeling mechanisms and microdamage dynamics determines its stiffness, strength, and toughness. Aging and disease affect the biological processes regulating bone quality, thus resulting in defective extracellular matrix and bone fragility. Targeted therapies are being developed to restore bone's mechanical integrity. However, their current limitations include low tissue selectivity and adverse side effects. Biological and mechanical insights into the mechanisms controlling bone quality, together with advances in drug delivery and studies in animal models, will accelerate the development and translation to clinical application of effective targeted-therapeutics for bone fragility.
Collapse
Affiliation(s)
- Asier Muñoz
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA
| | - Anxhela Docaj
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA
| | - Maialen Ugarteburu
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA
| | - Alessandra Carriero
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA.
| |
Collapse
|
20
|
Greene B, Russo RJ, Dwyer S, Malley K, Roberts E, Serrielo J, Piepenhagen P, Cummings S, Ryan S, Zarazinski C, Uppuganti S, Bukanov N, Nyman JS, Cox MK, Liu S, Ibraghimov-Beskrovnaya O, Sabbagh Y. Inhibition of TGF-β Increases Bone Volume and Strength in a Mouse Model of Osteogenesis Imperfecta. JBMR Plus 2021; 5:e10530. [PMID: 34532615 PMCID: PMC8441395 DOI: 10.1002/jbm4.10530] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/14/2021] [Accepted: 07/02/2021] [Indexed: 12/29/2022] Open
Abstract
Osteogenesis imperfecta (OI), is a genetic disorder of bone fragility caused by mutations in collagen I or proteins involved in collagen processing. Previous studies in mice and human OI bones have shown that excessive activation of TGF-β signaling plays an important role in dominant and recessive OI disease progression. Inhibition of TGF-β signaling with a murine pan-specific TGF-β neutralizing antibody (1D11) was shown to significantly increase trabecular bone volume and long bone strength in mouse models of OI. To investigate the frequency of dosing and dose options of TGF-β neutralizing antibody therapy, we assessed the effect of 1D11 on disease progression in a dominant OI mouse model (col1a2 gene mutation at G610C). In comparison with OI mice treated with a control antibody, we attempted to define mechanistic effects of 1D11 measured via μCT, biomechanical, dynamic histomorphometry, and serum biomarkers of bone turnover. In addition, osteoblast and osteoclast numbers in histological bone sections were assessed to better understand the mechanism of action of the 1D11 antibody in OI. Here we show that 1D11 treatment resulted in both dose and frequency dependency, increases in trabecular bone volume fraction and ultimate force in lumbar bone, and ultimate force, bending strength, yield force, and yield strength in the femur (p ≤ 0.05). Suppression of serum biomarkers of osteoblast differentiation, osteocalcin, resorption, CTx-1, and bone formation were observed after 1D11 treatment of OI mice. Immunohistochemical analysis showed dose and frequency dependent decreases in runt-related transcription factor, and increase in alkaline phosphatase in lumbar bone sections. In addition, a significant decrease in TRACP and the number of osteoclasts to bone surface area was observed with 1D11 treatment. Our results show that inhibition of the TGF-β pathway corrects the high-turnover aspects of bone disease and improves biomechanical properties of OI mice. These results highlight the potential for a novel treatment for osteogenesis imperfecta. © 2021 Sanofi-Genzyme. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Benjamin Greene
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | - Ryan J Russo
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | - Shannon Dwyer
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | - Katie Malley
- Global Discovery Pathology Sanofi Framingham MA USA
| | | | - Joseph Serrielo
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | | | | | - Susan Ryan
- Global Discovery Pathology Sanofi Framingham MA USA
| | | | - Sasidhar Uppuganti
- Department of Orthopaedic Surgery Vanderbilt University Medical Center Nashville TN USA.,Center for Bone Biology Vanderbilt University Medical Center Nashville TN USA
| | - Nikolai Bukanov
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | - Jeffry S Nyman
- Department of Orthopaedic Surgery Vanderbilt University Medical Center Nashville TN USA.,Center for Bone Biology Vanderbilt University Medical Center Nashville TN USA
| | - Megan K Cox
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | - Shiguang Liu
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA
| | | | - Yves Sabbagh
- Rare and Neurologic Diseases Research Sanofi Framingham MA USA.,Inozyme Pharma Boston MA USA
| |
Collapse
|
21
|
Yang H, Cao Z, Wang Y, Wang J, Gao J, Han B, Yu F, Qin Y, Guo Y. Treadmill exercise influences the microRNA profiles in the bone tissues of mice. Exp Ther Med 2021; 22:1035. [PMID: 34373721 PMCID: PMC8343800 DOI: 10.3892/etm.2021.10467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 05/28/2021] [Indexed: 12/12/2022] Open
Abstract
As an important regulator involved in cell activity, microRNAs (miRNAs) are important in the process of exercise influencing bone metabolism. The present study aimed to detect and select differentially expressed miRNAs in the bone tissues of mice trained on a treadmill, predict the target genes of these differentially expressed miRNAs and lay a foundation for exploring the effect of treadmill training on bone metabolism through miRNAs. In this experiment, after the mice were trained on a treadmill for 8 weeks, the mechanical properties of mouse femur bone were assessed, and the alkaline phosphatase (ALP) activity and osteocalcin (OCN) protein levels of the bone were assayed. miRNA microarray and reverse transcription-quantitative (RT-q)PCR were performed to select and validate differentially expressed miRNAs in the bone, and the target genes of these miRNAs were predicted with bioinformatics methods. In addition, the differentially expressed miRNAs in the bone tissues were compared with those in mechanically strained osteocytes in vitro. Treadmill training improved the mechanical properties of the femur bones of mice, and elevated the ALP activity and OCN protein level in the bone. In addition, 122 differentially expressed miRNAs were detected in the bone, of which nine were validated via RT-qPCR. Among the target genes of these differentially expressed miRNAs, certain candidates were involved in bone metabolism. A total of eight miRNAs were differentially expressed in both bone tissue and osteocytes, exhibiting the same expression trends, and various target genes of these eight miRNAs were also involved in bone metabolism. Treadmill training resulted in altered miRNA expression profiles in the bones of mice (mainly in osteocytes) and the differentially expressed miRNAs may serve important roles in regulating bone metabolism and osteogenic differentiation.
Collapse
Affiliation(s)
- Huan Yang
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Zhen Cao
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yang Wang
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China.,Department of Biomedical Engineering, Bioengineering College of Chongqing University, Chongqing 400044, P.R. China
| | - Jiahui Wang
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Jintao Gao
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Biao Han
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Fangmei Yu
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yixiong Qin
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| | - Yong Guo
- Department of Biomedical Engineering, College of Biotechnology, Guilin Medical University, Guilin, Guangxi 541004, P.R. China
| |
Collapse
|
22
|
Abstract
Osteogenesis imperfecta (OI) is a disease characterised by altered bone tissue material properties together with abnormal micro and macro-architecture and thus bone fragility, increased bone turnover and hyperosteocytosis. Increasingly appreciated are the soft tissue changes, sarcopenia in particular. Approaches to treatment are now multidisciplinary, with bisphosphonates having been the primary pharmacological intervention over the last 20 years. Whilst meta-analyses suggest that anti-fracture efficacy across the life course is equivocal, there is good evidence that for children bisphosphonates reduce fracture risk, increase vertebral size and improve vertebral shape, as well as improving motor function and mobility. The genetics of OI continues to provide insights into the molecular pathogenesis of the disease, although the pathophysiology is less clear. The complexity of the multi-scale interactions of bone tissue with cellular function are gradually being disentangled, but the fundamental question of why increased tissue brittleness should be associated with so many other changes is unclear; ER stress, pro-inflammatory cytokines, accelerated senesence and altered matrix component release might all contribute, but a unifying hypothesis remains elusive. New approaches to therapy are focussed on increasing bone mass, following the paradigm established by the treatment of postmenopausal osteoporosis. For adults, this brings the prospect of restoring previously lost bone - for children, particularly at the severe end of the spectrum, the possibility of further reducing fracture frequency and possibly altering growth and long term function are attractive. The alternatives that might affect tissue brittleness are autophagy enhancement (through the removal of abnormal type I collagen aggregates) and stem cell transplantation - both still at the preclinical stage of assessment. Preclinical assessment is not supportive of targeting inflammatory pathways, although understanding why TGFb signalling is increased, and whether that presents a treatment target in OI, remains to be established.
Collapse
Affiliation(s)
- Fawaz Arshad
- Academic Unit of Child Health, Sheffield Children's Hospital, Department of Oncology and Metabolism, University of Sheffield, S10 2TH, UK
| | - Nick Bishop
- Academic Unit of Child Health, Sheffield Children's Hospital, Department of Oncology and Metabolism, University of Sheffield, S10 2TH, UK.
| |
Collapse
|
23
|
Zhen G, Guo Q, Li Y, Wu C, Zhu S, Wang R, Guo XE, Kim BC, Huang J, Hu Y, Dan Y, Wan M, Ha T, An S, Cao X. Mechanical stress determines the configuration of TGFβ activation in articular cartilage. Nat Commun 2021; 12:1706. [PMID: 33731712 PMCID: PMC7969741 DOI: 10.1038/s41467-021-21948-0] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 02/19/2021] [Indexed: 01/18/2023] Open
Abstract
Our incomplete understanding of osteoarthritis (OA) pathogenesis has significantly hindered the development of disease-modifying therapy. The functional relationship between subchondral bone (SB) and articular cartilage (AC) is unclear. Here, we found that the changes of SB architecture altered the distribution of mechanical stress on AC. Importantly, the latter is well aligned with the pattern of transforming growth factor beta (TGFβ) activity in AC, which is essential in the regulation of AC homeostasis. Specifically, TGFβ activity is concentrated in the areas of AC with high mechanical stress. A high level of TGFβ disrupts the cartilage homeostasis and impairs the metabolic activity of chondrocytes. Mechanical stress stimulates talin-centered cytoskeletal reorganization and the consequent increase of cell contractile forces and cell stiffness of chondrocytes, which triggers αV integrin–mediated TGFβ activation. Knockout of αV integrin in chondrocytes reversed the alteration of TGFβ activation and subsequent metabolic abnormalities in AC and attenuated cartilage degeneration in an OA mouse model. Thus, SB structure determines the patterns of mechanical stress and the configuration of TGFβ activation in AC, which subsequently regulates chondrocyte metabolism and AC homeostasis. The functional relationship between subchondral bone and articular cartilage is unclear. Here, the authors show that transforming growth factor-beta propagates the mechanical impact of subchondral bone on articular cartilage through αV integrin–talin mechanical transduction system in chondrocytes.
Collapse
Affiliation(s)
- Gehua Zhen
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Qiaoyue Guo
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Yusheng Li
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Chuanlong Wu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Shouan Zhu
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Ruomei Wang
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - X Edward Guo
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Byoung Choul Kim
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University, Baltimore, MD, USA
| | - Jessie Huang
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ, USA
| | - Yizhong Hu
- Department of Biomedical Engineering, Columbia University, New York, NY, USA
| | - Yang Dan
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Mei Wan
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA
| | - Taekjip Ha
- Department of Biophysics and Biophysical Chemistry, The Johns Hopkins University, Baltimore, MD, USA
| | - Steven An
- Department of Pharmacology, Rutgers-Robert Wood Johnson Medical School, The State University of New Jersey, Piscataway, NJ, USA.,Rutgers Institute for Translational Medicine and Science, New Brunswick, NJ, USA
| | - Xu Cao
- Department of Orthopaedic Surgery, The Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
24
|
Bailey KN, Nguyen J, Yee CS, Dole NS, Dang A, Alliston T. Mechanosensitive Control of Articular Cartilage and Subchondral Bone Homeostasis in Mice Requires Osteocytic Transforming Growth Factor β Signaling. Arthritis Rheumatol 2021; 73:414-425. [PMID: 33022131 DOI: 10.1002/art.41548] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 09/23/2020] [Indexed: 12/25/2022]
Abstract
OBJECTIVE Transforming growth factor β (TGFβ) signaling plays a complex tissue-specific and nonlinear role in osteoarthritis (OA). This study was conducted to determine the osteocytic contributions of TGFβ signaling to OA. METHODS To identify the role of osteocytic TGFβ signaling in joint homeostasis, we used 16-week-old male mice (n = 9-11 per group) and female mice (n = 7-11 per group) with an osteocyte-intrinsic ablation of TGFβ receptor type II (TβRIIocy-/- mice) and assessed defects in cartilage degeneration, subchondral bone plate (SBP) thickness, and SBP sclerostin expression. To further investigate these mechanisms in 16-week-old male mice, we perturbed joint homeostasis by subjecting 8-week-old mice to medial meniscal/ligamentous injury (MLI), which preferentially disrupts the mechanical environment of the medial joint to induce OA. RESULTS In all contexts, independent of sex, genotype, or medial or lateral joint compartment, increased SBP thickness and SBP sclerostin expression were spatially associated with cartilage degeneration. Male TβRIIocy-/- mice, but not female TβRIIocy-/- mice, had increased cartilage degeneration, increased SBP thickness, and higher levels of SBP sclerostin compared with control mice (all P < 0.05), demonstrating that the role of osteocytic TGFβ signaling on joint homeostasis is sexually dimorphic. With changes in joint mechanics following injury, control mice had increased SBP thickness, subchondral bone volume, and SBP sclerostin expression (all P < 0.05). TβRIIocy-/- mice, however, were insensitive to subchondral bone changes with injury, suggesting that mechanosensation at the SBP requires osteocytic TGFβ signaling. CONCLUSION Our results provide new evidence that osteocytic TGFβ signaling is required for a mechanosensitive response to injury, and that osteocytes control SBP homeostasis to maintain cartilage health, identifying osteocytic TGFβ signaling as a novel therapeutic target for OA.
Collapse
Affiliation(s)
| | - Jeffrey Nguyen
- University of California, San Francisco, and California State University, Long Beach
| | | | | | - Alexis Dang
- University of California, San Francisco and San Francisco VAMC, San Francisco, California
| | | |
Collapse
|
25
|
Siddiqui JA, Le Henaff C, Johnson J, He Z, Rifkin DB, Partridge NC. Osteoblastic monocyte chemoattractant protein-1 (MCP-1) mediation of parathyroid hormone's anabolic actions in bone implicates TGF-β signaling. Bone 2021; 143:115762. [PMID: 33212319 PMCID: PMC8628523 DOI: 10.1016/j.bone.2020.115762] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 11/12/2020] [Accepted: 11/14/2020] [Indexed: 12/24/2022]
Abstract
Parathyroid hormone (PTH) is necessary for the regulation of calcium homeostasis and PTH (1-34) was the first approved osteoanabolic therapy for osteoporosis. It is well established that intermittent PTH increases bone formation and that bone remodeling and several cytokines and chemokines play an essential role in this process. Earlier, we had established that the chemokine, monocyte chemoattractant protein-1 (MCP-1/CCL2), was the most highly stimulated gene in rat bone after intermittent PTH injections. Nevertheless, MCP-1 function in bone appears to be complicated. To identify the primary cells expressing MCP-1 in response to PTH, we performed in situ hybridization of rat bone sections after hPTH (1-34) injections and showed that bone-lining osteoblasts are the primary cells that express MCP-1 after PTH treatment. We previously demonstrated MCP-1's importance by showing that PTH's anabolic effects are abolished in MCP-1 null mice, further implicating a role for the chemokine in this process. To establish whether rhMCP-1 peptide treatment could rescue the anabolic effect of PTH in MCP-1 null mice, we treated 4-month-old wild-type (WT) mice with hPTH (1-34) and MCP-1-/- mice with rhMCP-1 and/or hPTH (1-34) for 6 weeks. Micro-computed tomography (μCT) analysis of trabecular and cortical bone showed that MCP-1 injections for 6 weeks rescued the PTH anabolic effect in MCP-1-/- mice. In fact, the combination of rhMCP-1 and hPTH (1-34) has a synergistic anabolic effect compared with monotherapies. Mechanistically, PTH-enhanced transforming growth factor-β (TGF-β) signaling is abolished in the absence of MCP-1, while MCP-1 peptide treatment restores TGF-β signaling in the bone marrow. Here, we have shown that PTH regulates the transcription of the chemokine MCP-1 in osteoblasts and determined how MCP-1 affects bone cell function in PTH's anabolic actions. Taken together, our current work indicates that intermittent PTH stimulates osteoblastic secretion of MCP-1, which leads to increased TGF-β signaling, implicating it in PTH's anabolic actions.
Collapse
Affiliation(s)
- Jawed A Siddiqui
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, United States of America
| | - Carole Le Henaff
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, United States of America
| | - Joshua Johnson
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, United States of America
| | - Zhiming He
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, United States of America
| | - Daniel B Rifkin
- Department of Cell Biology, New York University Grossman School of Medicine, New York, United States of America
| | - Nicola C Partridge
- Department of Molecular Pathobiology, New York University College of Dentistry, New York, United States of America.
| |
Collapse
|
26
|
Teodorescu P, Pasca S, Jurj A, Gafencu G, Joelsson JP, Selicean S, Moldovan C, Munteanu R, Onaciu A, Tigu AB, Buse M, Zimta AA, Stiufiuc R, Petrushev B, Desmirean M, Dima D, Vlad C, Bergthorsson JT, Berce C, Ciurea S, Ghiaur G, Tomuleasa C. Transforming growth factor β-mediated micromechanics modulates disease progression in primary myelofibrosis. J Cell Mol Med 2020; 24:11100-11110. [PMID: 32889753 PMCID: PMC7576271 DOI: 10.1111/jcmm.15526] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 05/29/2020] [Indexed: 12/15/2022] Open
Abstract
Primary myelofibrosis (PMF) is a Ph‐negative myeloproliferative neoplasm (MPN), characterized by advanced bone marrow fibrosis and extramedullary haematopoiesis. The bone marrow fibrosis results from excessive proliferation of fibroblasts that are influenced by several cytokines in the microenvironment, of which transforming growth factor‐β (TGF‐β) is the most important. Micromechanics related to the niche has not yet been elucidated. In this study, we hypothesized that mechanical stress modulates TGF‐β signalling leading to further activation and subsequent proliferation and invasion of bone marrow fibroblasts, thus showing the important role of micromechanics in the development and progression of PMF, both in the bone marrow and in extramedullary sites. Using three PMF‐derived fibroblast cell lines and transforming growth factor‐β receptor (TGFBR) 1 and 2 knock‐down PMF‐derived fibroblasts, we showed that mechanical stress does stimulate the collagen synthesis by the fibroblasts in patients with myelofibrosis, through the TGFBR1, which however seems to be activated through alternative pathways, other than TGFBR2.
Collapse
Affiliation(s)
- Patric Teodorescu
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania
| | - Sergiu Pasca
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Ancuta Jurj
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Grigore Gafencu
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Molecular Haematology Unit - Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jon-Petur Joelsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland.,Department of Laboratory Hematology, Landspitali University Hospital, Reykjavík, Iceland
| | - Sonia Selicean
- Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Graduate School for Cellular and Biomedical Sciences, Universität Bern, Bern, Switzerland
| | - Cristian Moldovan
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Raluca Munteanu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Anca Onaciu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Adrian-Bogdan Tigu
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Mihail Buse
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Alina-Andreea Zimta
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Rares Stiufiuc
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Bobe Petrushev
- Medfuture Research Center for Advanced Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Minodora Desmirean
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Pathology, Constantin Papilian Military Hospital, Cluj Napoca, Romania
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Clinical Research Center, Cluj Napoca, Romania
| | - Cristina Vlad
- Department of Cardiology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Cardiology, Rehabilitation Hospital, Cluj Napoca, Romania
| | - Jon Thor Bergthorsson
- Stem Cell Research Unit, Biomedical Center, School of Health Sciences, University of Iceland, Reykjavík, Iceland.,Department of Laboratory Hematology, Landspitali, University Hospital, Reykjavík, Iceland
| | - Cristian Berce
- Animal Facility, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| | - Stefan Ciurea
- Department of Cellular Therapies and Stem Cell Transplantation, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gabriel Ghiaur
- Department of Leukemia, Sidney Kimmel Cancer Center at Johns Hopkins, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ciprian Tomuleasa
- Department of Hematology, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania.,Department of Hematology, Ion Chiricuta Clinical Cancer Center, Cluj Napoca, Romania.,Research Center for Functional Genomics and Translational Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, Cluj Napoca, Romania
| |
Collapse
|
27
|
Bolger MW, Romanowicz GE, Kohn DH. Advancements in composition and structural characterization of bone to inform mechanical outcomes and modelling. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2020; 11:76-84. [PMID: 32864522 DOI: 10.1016/j.cobme.2019.09.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Advancements in imaging, computing, microscopy, chromatography, spectroscopy and biological manipulations of animal models, have allowed for a more thorough examination of the hierarchical structure and composition of the skeleton. The ability to map cellular and molecular changes to nano-scale chemical composition changes (mineral, collagen cross-links) and structural changes (porosity, lacuno-canalicular network) to whole bone mechanics is at the forefront of an exciting era of discovery. In addition, there is increasing ability to genetically mimic phenotypes of human disease in animal models to study these structural and compositional changes. Combined, these recent developments have increased the ability to understand perturbations at multiple length scales to better realize the structure-function relationship in bone and inform biomechanical models. The intent of this review is to describe the multiple scales at which bone can characterized, highlighting new techniques such that structural, compositional, and biological changes can be incorporated into biomechanical modeling.
Collapse
Affiliation(s)
- Morgan W Bolger
- Biomedical Engineering, College of Engineering, University of Michigan, MI, USA
| | - Genevieve E Romanowicz
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| | - David H Kohn
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, MI, USA
| |
Collapse
|
28
|
Trifirò G, Mora S, Marelli S, Luzi L, Pini A. Increased fracture rate in children and adolescents with Marfan syndrome. Bone 2020; 135:115333. [PMID: 32222606 DOI: 10.1016/j.bone.2020.115333] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/17/2020] [Accepted: 03/24/2020] [Indexed: 11/26/2022]
Abstract
Marfan syndrome (MFS) is an autosomal genetic disorder of connective tissue, due to alterated fibrillin-1. The aim of our study was to verify the rate of fractures in children with MFS in correlation to bone mineral density and compare the prevalence to the general population in the same latitude. We enrolled 80 patients (37 girls and 43 boys) with the diagnosis of Marfan syndrome, median age 10 y (3 to 17 years). Fracture occurrence was inferred from medical records of patients with MFS. Bone mineral density (BMD) was measured at lumbar spine, femoral neck and total femur by dual-energy x-ray absorptiometry. BMD values were expressed as z-scores, and adjusted for height using height-for-age z-scores. Bone turnover markers and vitamin D were measured. We assessed incidence of fracture in general pediatric population of our geographic area (45°N latitude). A total of 24 fractures were recorded in 21 patients (15 boys and 6 girls), involving both short and long bones, due to mild or moderate trauma. An incidence estimate has been calculated for each year, and an average incidence of 29.2/1000 MFS patients was obtained, markedly higher (P=0.034) than the incidence of fracture calculated in the same geographical area in pediatric patients (15.8/1000). We did not detect differences in anthropometric measurements, BMD values and biochemical indices between patients who fractured and patients who did not. Similarly, no differences were found between patients on losartan therapy and patients not in treatment for the same variables. In conclusion, the incidence of fractures was higher in patients with MFS compared to general population of the same age and latitude. The management of MFS must account bone status health and start strategies of fracture prevention.
Collapse
Affiliation(s)
- Giuliana Trifirò
- Endocrinology and Metabolism Division, IRCCS Policlinico San Donato, Milan, Italy.
| | - Stefano Mora
- Laboratory of Pediatric Endocrinology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Susan Marelli
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, Milan, Italy
| | - Livio Luzi
- Endocrinology and Metabolism Division and Università degli Studi di Milano, IRCCS Policlinico San Donato, Milan, Italy
| | - Alessandro Pini
- Cardiovascular-Genetic Center, IRCCS Policlinico San Donato, Milan, Italy
| |
Collapse
|
29
|
Hardy E, Fernandez-Patron C. Destroy to Rebuild: The Connection Between Bone Tissue Remodeling and Matrix Metalloproteinases. Front Physiol 2020; 11:47. [PMID: 32116759 PMCID: PMC7013034 DOI: 10.3389/fphys.2020.00047] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 01/21/2020] [Indexed: 12/11/2022] Open
Abstract
Bone is a dynamic organ that undergoes constant remodeling, an energetically costly process by which old bone is replaced and localized bone defects are repaired to renew the skeleton over time, thereby maintaining skeletal health. This review provides a general overview of bone’s main players (bone lining cells, osteocytes, osteoclasts, reversal cells, and osteoblasts) that participate in bone remodeling. Placing emphasis on the family of extracellular matrix metalloproteinases (MMPs), we describe how: (i) Convergence of multiple protease families (including MMPs and cysteine proteinases) ensures complexity and robustness of the bone remodeling process, (ii) Enzymatic activity of MMPs affects bone physiology at the molecular and cellular levels and (iii) Either overexpression or deficiency/insufficiency of individual MMPs impairs healthy bone remodeling and systemic metabolism. Today, it is generally accepted that proteolytic activity is required for the degradation of bone tissue in osteoarthritis and osteoporosis. However, it is increasingly evident that inactivating mutations in MMP genes can also lead to bone pathology including osteolysis and metabolic abnormalities such as delayed growth. We argue that there remains a need to rethink the role played by proteases in bone physiology and pathology.
Collapse
Affiliation(s)
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
30
|
Treurniet S, Eekhoff EMW, Schmidt FN, Micha D, Busse B, Bravenboer N. A Clinical Perspective on Advanced Developments in Bone Biopsy Assessment in Rare Bone Disorders. Front Endocrinol (Lausanne) 2020; 11:399. [PMID: 32714279 PMCID: PMC7344330 DOI: 10.3389/fendo.2020.00399] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 05/18/2020] [Indexed: 01/22/2023] Open
Abstract
Introduction: Bone biopsies have been obtained for many centuries and are one of the oldest known medical procedures in history. Despite the introduction of new noninvasive radiographic imaging techniques and genetic analyses, bone biopsies are still valuable in the diagnosis of bone diseases. Advanced techniques for the assessment of bone quality in bone biopsies, which have emerged during the last decades, allows in-depth tissue analyses beyond structural changes visible in bone histology. In this review, we give an overview of the application and advantages of the advanced techniques for the analysis of bone biopsies in the clinical setting of various rare metabolic bone diseases. Method: A systematic literature search on rare metabolic bone diseases and analyzing techniques of bone biopsies was performed in PubMed up to 2019 week 34. Results: Advanced techniques for the analysis of bone biopsies were described for rare metabolic bone disorders including Paget's disease of bone, osteogenesis imperfecta, fibrous dysplasia, Fibrodysplasia ossificans progressiva, PLS3 X-linked osteoporosis, Loeys-Diets syndrome, osteopetrosis, Erdheim-Chester disease, and Cherubism. A variety of advanced available analytical techniques were identified that may help to provide additional detail on cellular, structural, and compositional characteristics in rare bone diseases complementing classical histopathology. Discussion: To date, these techniques have only been used in research and not in daily clinical practice. Clinical application of bone quality assessment techniques depends upon several aspects such as availability of the technique in hospitals, the existence of reference data, and a cooperative network of researchers and clinicians. The evaluation of rare metabolic bone disorders requires a repertoire of different methods, owing to their distinct bone tissue characteristics. The broader use of bone material obtained from biopsies could provide much more information about pathophysiology or treatment options and establish bone biopsies as a valuable tool in rare metabolic bone diseases.
Collapse
Affiliation(s)
- Sanne Treurniet
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Elisabeth M. W. Eekhoff
- Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam Movement Sciences, Amsterdam, Netherlands
| | - Felix N. Schmidt
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dimitra Micha
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Nathalie Bravenboer
- Bone and Calcium Metabolism Lab, Department of Clinical Chemistry, Amsterdam University Medical Center, Amsterdam Movement Sciences, Amsterdam, Netherlands
- *Correspondence: Nathalie Bravenboer
| |
Collapse
|
31
|
Li B, Fang L, Null DJ, Hutchison JL, Connor EE, VanRaden PM, VandeHaar MJ, Tempelman RJ, Weigel KA, Cole JB. High-density genome-wide association study for residual feed intake in Holstein dairy cattle. J Dairy Sci 2019; 102:11067-11080. [PMID: 31563317 DOI: 10.3168/jds.2019-16645] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/19/2019] [Indexed: 01/27/2023]
Abstract
Improving feed efficiency (FE) of dairy cattle may boost farm profitability and reduce the environmental footprint of the dairy industry. Residual feed intake (RFI), a candidate FE trait in dairy cattle, can be defined to be genetically uncorrelated with major energy sink traits (e.g., milk production, body weight) by including genomic predicted transmitting ability of such traits in genetic analyses for RFI. We examined the genetic basis of RFI through genome-wide association (GWA) analyses and post-GWA enrichment analyses and identified candidate genes and biological pathways associated with RFI in dairy cattle. Data were collected from 4,823 lactations of 3,947 Holstein cows in 9 research herds in the United States. Of these cows, 3,555 were genotyped and were imputed to a high-density list of 312,614 SNP. We used a single-step GWA method to combine information from genotyped and nongenotyped animals with phenotypes as well as their ancestors' information. The estimated genomic breeding values from a single-step genomic BLUP were back-solved to obtain the individual SNP effects for RFI. The proportion of genetic variance explained by each 5-SNP sliding window was also calculated for RFI. Our GWA analyses suggested that RFI is a highly polygenic trait regulated by many genes with small effects. The closest genes to the top SNP and sliding windows were associated with dry matter intake (DMI), RFI, energy homeostasis and energy balance regulation, digestion and metabolism of carbohydrates and proteins, immune regulation, leptin signaling, mitochondrial ATP activities, rumen development, skeletal muscle development, and spermatogenesis. The region of 40.7 to 41.5 Mb on BTA25 (UMD3.1 reference genome) was the top associated region for RFI. The closest genes to this region, CARD11 and EIF3B, were previously shown to be related to RFI of dairy cattle and FE of broilers, respectively. Another candidate region, 57.7 to 58.2 Mb on BTA18, which is associated with DMI and leptin signaling, was also associated with RFI in this study. Post-GWA enrichment analyses used a sum-based marker-set test based on 4 public annotation databases: Gene Ontology, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways, Reactome pathways, and medical subject heading (MeSH) terms. Results of these analyses were consistent with those from the top GWA signals. Across the 4 databases, GWA signals for RFI were highly enriched in the biosynthesis and metabolism of amino acids and proteins, digestion and metabolism of carbohydrates, skeletal development, mitochondrial electron transport, immunity, rumen bacteria activities, and sperm motility. Our findings offer novel insight into the genetic basis of RFI and identify candidate regions and biological pathways associated with RFI in dairy cattle.
Collapse
Affiliation(s)
- B Li
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350
| | - L Fang
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350; Department of Animal and Avian Sciences, University of Maryland, College Park 20742; Medical Research Council Human Genetics Unit at the Medical Research Council Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, EH4 2XU, United Kingdom
| | - D J Null
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350
| | - J L Hutchison
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350
| | - E E Connor
- Department of Animal and Food Sciences, University of Delaware, Newark 19716
| | - P M VanRaden
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350
| | - M J VandeHaar
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - R J Tempelman
- Department of Animal Science, Michigan State University, East Lansing 48824
| | - K A Weigel
- Department of Dairy Science, University of Wisconsin, Madison 53706
| | - J B Cole
- Animal Genomics and Improvement Laboratory, Agricultural Research Service, USDA, Beltsville, MD 20705-2350.
| |
Collapse
|
32
|
Yan XZ, van den Beucken JJJP, Yuan C, Jansen JA, Yang F. Evaluation of polydimethylsiloxane-based substrates for in vitro culture of human periodontal ligament cells. J Biomed Mater Res A 2019; 107:2796-2805. [PMID: 31408269 DOI: 10.1002/jbm.a.36782] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/05/2019] [Accepted: 08/07/2019] [Indexed: 12/11/2022]
Abstract
Periodontal ligament (PDL) cells are regarded as the cell type with the highest potential for periodontal regeneration. Biophysical cues of the culture substrate are increasingly identified as vital parameters to affect cell behavior. Compared to traditional tissue culture polystyrene (TCPS), polydimethylsiloxane (PDMS) substrates corroborate more closely the elastic modulus values of the physiological environment. Consequently, the aim of this study was to evaluate the effect of PDMS-based substrates with different stiffness on cellular responses of human PDL cells. PDMS substrates with different stiffness were fabricated by varying the ratio of base to curing component. The influence of PDMS substrates on PDL cell spreading and cytoskeletal morphologies, motility, proliferation, stemness gene expression, and osteogenic differentiation was evaluated and compared to that on conventional TCPS. PDL cells cultured on PDMS substrates exhibited a smaller cell size and more elongated morphology, with less spreading area, fewer focal adhesions, and faster migration than cells on TCPS. Compared to TCPS, PDMS substrates promoted the rapid in vitro expansion of PDL cells without interfering with their self-renewal ability. In contrast, the osteogenic differentiation ability of PDL cells cultured on PDMS was lower in comparison to cells on TCPS. PDL cells on PDMS exhibited similar cell morphology, motility, proliferation, and self-renewal gene expression. The stiffer PDMS substrate increased the osteogenic gene expression of PDL cells compared to the soft PDMS group in one donor. These data indicate that PDMS-based substrates have the potential for the efficient PDL cell expansion.
Collapse
Affiliation(s)
- Xiang-Zhen Yan
- Department of Periodontology, School and hospital of Stomatology, Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Shanghai, China
| | | | - Chunxue Yuan
- College of Materials Science and Engineering, Tongji University, Shanghai, China
| | - John A Jansen
- Department of Dentistry - Biomaterials, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Fang Yang
- Department of Dentistry - Biomaterials, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
33
|
Betaglycan drives the mesenchymal stromal cell osteogenic program and prostate cancer-induced osteogenesis. Oncogene 2019; 38:6959-6969. [PMID: 31409900 DOI: 10.1038/s41388-019-0913-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 07/04/2019] [Accepted: 07/10/2019] [Indexed: 12/11/2022]
Abstract
Bone metastatic prostate cancer provokes extensive osteogenesis by driving the recruitment and osteoblastic differentiation of mesenchymal stromal cells (MSCs). The resulting lesions greatly contribute to patient morbidity and mortality, underscoring the need for defining how prostate metastases subvert the MSC-osteoblast differentiation program. To gain insights into this process we profiled the effects of co-culture of primary MSCs with validated bone metastatic prostate cancer cell line models. These analyses revealed a cast of shared differentially induced genes in MSC, including betaglycan, a co-receptor for TGFβ. Betaglycan has not been studied in the context of bone metastatic disease previously. Here we report that loss of betaglycan in MSC is sufficient to augment TGFβ signaling, proliferation and migration, and completely blocks the MSC-osteoblast differentiation program. Further, betaglycan was revealed as necessary for prostate cancer-induced osteogenesis in vivo. Mechanistically, gene expression analysis revealed betaglycan controls the expression of a large repertoire of genes in MSCs, and that betaglycan loss provokes >60-fold increase in the expression of Wnt5a that plays important roles in stemness. In accord with the increased Wnt5a levels, there was a marked induction of canonical Wnt signaling in betaglycan ablated MSCs, and the addition of recombinant Wnt5a to MSCs was sufficient to impair osteogenic differentiation. Finally, the addition of Wnt5a neutralizing antibody was sufficient to induce the expression of osteogenic genes in betaglycan-ablated MSCs. Collectively, these findings suggest a betaglycan-Wnt5a circuit represents an attractive vulnerability to ameliorate prostate cancer-induced osteogenesis.
Collapse
|
34
|
Woronowicz KC, Schneider RA. Molecular and cellular mechanisms underlying the evolution of form and function in the amniote jaw. EvoDevo 2019; 10:17. [PMID: 31417668 PMCID: PMC6691539 DOI: 10.1186/s13227-019-0131-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Accepted: 07/22/2019] [Indexed: 01/16/2023] Open
Abstract
The amniote jaw complex is a remarkable amalgamation of derivatives from distinct embryonic cell lineages. During development, the cells in these lineages experience concerted movements, migrations, and signaling interactions that take them from their initial origins to their final destinations and imbue their derivatives with aspects of form including their axial orientation, anatomical identity, size, and shape. Perturbations along the way can produce defects and disease, but also generate the variation necessary for jaw evolution and adaptation. We focus on molecular and cellular mechanisms that regulate form in the amniote jaw complex, and that enable structural and functional integration. Special emphasis is placed on the role of cranial neural crest mesenchyme (NCM) during the species-specific patterning of bone, cartilage, tendon, muscle, and other jaw tissues. We also address the effects of biomechanical forces during jaw development and discuss ways in which certain molecular and cellular responses add adaptive and evolutionary plasticity to jaw morphology. Overall, we highlight how variation in molecular and cellular programs can promote the phenomenal diversity and functional morphology achieved during amniote jaw evolution or lead to the range of jaw defects and disease that affect the human condition.
Collapse
Affiliation(s)
- Katherine C Woronowicz
- 1Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1161, Box 0514, San Francisco, CA 94143-0514 USA.,2Present Address: Department of Genetics, Harvard Medical School, Orthopaedic Research Laboratories, Children's Hospital Boston, Boston, MA 02115 USA
| | - Richard A Schneider
- 1Department of Orthopaedic Surgery, University of California at San Francisco, 513 Parnassus Avenue, S-1161, Box 0514, San Francisco, CA 94143-0514 USA
| |
Collapse
|
35
|
Levels of Cytokines in Gingival Crevicular Fluid during Rapid Maxillary Expansion and the Subsequent Retention Period. J Clin Pediatr Dent 2019; 43:137-143. [PMID: 30730797 DOI: 10.17796/1053-4625-43.2.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
OBJECTIVE To monitor the effects of rapid maxillary expansion (RME) on bone metabolic activities during and after 3 months of retention. STUDY DESIGN Fifteen patients with a mean age of 12.9 ± 0.6 years were treated with a bonded expansion device, activated 2 turns per day. The retention period was 3 months. Clinical periodontal parameters were recorded at baseline and after retention. Gingival crevicular fluid (GCF) samples were collected from maxillary first molars from the compression sides at baseline, then at 1 and 10 days and after retention. Tension side samples were obtained at baseline and after retention. Interleukin-1beta (IL-1β), transforming growth factor beta1 (TGF-β1), prostaglandin E2 (PGE2) and nitric oxide (NO) levels were specifically measured. RESULTS Periodontal parameters increased significantly after retention relative to baseline values. Levels of IL-1β, TGF-β1 and PGE2 increased on day 10, and decreased after retention on the compression side. NO levels were elevated on day 10, and remained higher after retention on the compression side. Tension side cytokine levels remained higher relative to baseline values after retention. CONCLUSIONS The results of this study indicate the importance of ongoing adaptive bone activities after 3 months of retention with RME, which should be considered questionable as an effective retention period.
Collapse
|
36
|
Ishikawa J, Kano F, Ando Y, Hibi H, Yamamoto A. Monocyte chemoattractant protein-1 and secreted ectodomain of sialic acid-binding Ig-like lectin-9 enhance bone regeneration by inducing M2 macrophages. JOURNAL OF ORAL AND MAXILLOFACIAL SURGERY, MEDICINE, AND PATHOLOGY 2019. [DOI: 10.1016/j.ajoms.2018.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
37
|
Sun Y, Vu LH, Chew N, Puthucheary Z, Cove ME, Zeng K. A Study of Perturbations in Structure and Elastic Modulus of Bone Microconstituents Using Bimodal Amplitude Modulated-Frequency Modulated Atomic Force Microscopy. ACS Biomater Sci Eng 2018; 5:478-486. [DOI: 10.1021/acsbiomaterials.8b01087] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Yao Sun
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, 117576, Singapore
| | - Lien Hong Vu
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228
| | - Nicholas Chew
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228
| | - Zudin Puthucheary
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228
- Division of Critical Care, Institute of Sports and Exercise Health, University College London Hospitals, U.K., and Centre for Human Health and Performance, University College London, London WC1E 6BT, United Kingdom
| | - Matthew E. Cove
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block Level 10, Singapore 119228
| | - Kaiyang Zeng
- Department of Mechanical Engineering, National University of Singapore, 9 Engineering Drive 1, 117576, Singapore
| |
Collapse
|
38
|
Woronowicz KC, Gline SE, Herfat ST, Fields AJ, Schneider RA. FGF and TGFβ signaling link form and function during jaw development and evolution. Dev Biol 2018; 444 Suppl 1:S219-S236. [PMID: 29753626 PMCID: PMC6239991 DOI: 10.1016/j.ydbio.2018.05.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 04/20/2018] [Accepted: 05/06/2018] [Indexed: 12/14/2022]
Abstract
How does form arise during development and change during evolution? How does form relate to function, and what enables embryonic structures to presage their later use in adults? To address these questions, we leverage the distinct functional morphology of the jaw in duck, chick, and quail. In connection with their specialized mode of feeding, duck develop a secondary cartilage at the tendon insertion of their jaw adductor muscle on the mandible. An equivalent cartilage is absent in chick and quail. We hypothesize that species-specific jaw architecture and mechanical forces promote secondary cartilage in duck through the differential regulation of FGF and TGFβ signaling. First, we perform transplants between chick and duck embryos and demonstrate that the ability of neural crest mesenchyme (NCM) to direct the species-specific insertion of muscle and the formation of secondary cartilage depends upon the amount and spatial distribution of NCM-derived connective tissues. Second, we quantify motility and build finite element models of the jaw complex in duck and quail, which reveals a link between species-specific jaw architecture and the predicted mechanical force environment. Third, we investigate the extent to which mechanical load mediates FGF and TGFβ signaling in the duck jaw adductor insertion, and discover that both pathways are mechano-responsive and required for secondary cartilage formation. Additionally, we find that FGF and TGFβ signaling can also induce secondary cartilage in the absence of mechanical force or in the adductor insertion of quail embryos. Thus, our results provide novel insights on molecular, cellular, and biomechanical mechanisms that couple musculoskeletal form and function during development and evolution.
Collapse
Affiliation(s)
- Katherine C Woronowicz
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Stephanie E Gline
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Safa T Herfat
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Aaron J Fields
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA
| | - Richard A Schneider
- Department of Orthopaedic Surgery, University of California, San Francisco, 513 Parnassus Avenue, S-1161, San Francisco, CA 94143-0514, USA.
| |
Collapse
|
39
|
Ripamonti U. Developmental pathways of periodontal tissue regeneration. J Periodontal Res 2018; 54:10-26. [DOI: 10.1111/jre.12596] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 07/06/2018] [Accepted: 07/12/2018] [Indexed: 12/13/2022]
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory; School of Oral Health Sciences; Faculty of Health Sciences; University of the Witwatersrand; Johannesburg South Africa
| |
Collapse
|
40
|
Osteocyte-Intrinsic TGF-β Signaling Regulates Bone Quality through Perilacunar/Canalicular Remodeling. Cell Rep 2018; 21:2585-2596. [PMID: 29186693 DOI: 10.1016/j.celrep.2017.10.115] [Citation(s) in RCA: 110] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2017] [Revised: 09/26/2017] [Accepted: 10/29/2017] [Indexed: 02/08/2023] Open
Abstract
Poor bone quality contributes to bone fragility in diabetes, aging, and osteogenesis imperfecta. However, the mechanisms controlling bone quality are not well understood, contributing to the current lack of strategies to diagnose or treat bone quality deficits. Transforming growth factor beta (TGF-β) signaling is a crucial mechanism known to regulate the material quality of bone, but its cellular target in this regulation is unknown. Studies showing that osteocytes directly remodel their perilacunar/canalicular matrix led us to hypothesize that TGF-β controls bone quality through perilacunar/canalicular remodeling (PLR). Using inhibitors and mice with an osteocyte-intrinsic defect in TGF-β signaling (TβRIIocy-/-), we show that TGF-β regulates PLR in a cell-intrinsic manner to control bone quality. Altogether, this study emphasizes that osteocytes are key in executing the biological control of bone quality through PLR, thereby highlighting the fundamental role of osteocyte-mediated PLR in bone homeostasis and fragility.
Collapse
|
41
|
Betz VM, Kochanek S, Rammelt S, Müller PE, Betz OB, Messmer C. Recent advances in gene-enhanced bone tissue engineering. J Gene Med 2018; 20:e3018. [PMID: 29601661 DOI: 10.1002/jgm.3018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/18/2018] [Accepted: 03/18/2018] [Indexed: 12/13/2022] Open
Abstract
The loss of bone tissue represents a critical clinical condition that is frequently faced by surgeons. Substantial progress has been made in the area of bone research, providing insight into the biology of bone under physiological and pathological conditions, as well as tools for the stimulation of bone regeneration. The present review discusses recent advances in the field of gene-enhanced bone tissue engineering. Gene transfer strategies have emerged as highly effective tissue engineering approaches for supporting the repair of the musculoskeletal system. By contrast to treatment with recombinant proteins, genetically engineered cells can release growth factors at the site of injury over extended periods of time. Of particular interest are the expedited technologies that can be applied during a single surgical procedure in a cost-effective manner, allowing translation from bench to bedside. Several promising methods based on the intra-operative genetic manipulation of autologous cells or tissue fragments have been developed in preclinical studies. Moreover, gene therapy for bone regeneration has entered the clinical stage with clinical trials for the repair of alveolar bone. Current trends in gene-enhanced bone engineering are also discussed with respect to the movement of the field towards expedited, translational approaches. It is possible that gene-enhanced bone tissue engineering will become a clinical reality within the next few years.
Collapse
Affiliation(s)
- Volker M Betz
- Department of Gene Therapy, University of Ulm, Ulm, Germany.,Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| | | | - Stefan Rammelt
- University Center of Orthopedics and Traumatology and Center for Translational Bone, Joint and Soft Tissue Research, University Hospital Carl Gustav Carus Dresden, Technical University Dresden, Dresden, Germany
| | - Peter E Müller
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Oliver B Betz
- Department of Orthopedic Surgery, Physical Medicine and Rehabilitation, University Hospital Grosshadern, Ludwig-Maximilians-University Munich, Munich, Germany.,Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Carolin Messmer
- Center for Rehabilitation, RKU - University and Rehabilitation Hospitals Ulm, Ulm, Germany
| |
Collapse
|
42
|
Pei YF, Hu WZ, Yan MW, Li CW, Liu L, Yang XL, Hai R, Wang XY, Shen H, Tian Q, Deng HW, Zhang L. Joint study of two genome-wide association meta-analyses identified 20p12.1 and 20q13.33 for bone mineral density. Bone 2018; 110:378-385. [PMID: 29499414 PMCID: PMC6329308 DOI: 10.1016/j.bone.2018.02.027] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/23/2018] [Accepted: 02/26/2018] [Indexed: 01/19/2023]
Abstract
In the present study, aiming to identify loci associated with osteoporosis, we conducted a joint association study of 2 independent genome-wide association meta-analyses of femoral neck and lumbar spine bone mineral densities (BMDs): 1) an in-house study of 6 samples involving 7484 subjects, and 2) the GEFOS-seq study of 7 samples involving 32,965 subjects. The in-house samples were imputed by the 1000 genomes project phase 3 reference panel. SNP-based association test was applied to 7,998,108 autosomal SNPs in each meta-analysis, and for each SNP the 2 association signals were then combined for joint analysis and for mutual replication. Combining the evidence from both studies, we identified 2 novel loci associated with BMDs at the genome-wide significance level (α=5.0×10-8): 20p12.1 (rs73100693 p=2.65×10-8, closest gene MACROD2) and 20q13.33 (rs2380128 p=3.44×10-8, OSBPL2). We also replicated 7 loci that were reported by two recent studies on heel and total body BMD. Our findings provide useful insights that enhance our understanding of bone development, osteoporosis and fracture pathogenesis.
Collapse
Affiliation(s)
- Yu-Fang Pei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China; Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Wen-Zhu Hu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Min-Wei Yan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, PR China
| | - Chang-Wei Li
- Department of Epidemiology and Biostatistics, University of Georgia College of Public Health, Athens, GA, USA
| | - Lu Liu
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Xiao-Lin Yang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China
| | - Rong Hai
- Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, PR China
| | - Xiu-Yan Wang
- Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, PR China
| | - Hui Shen
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Qing Tian
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA
| | - Hong-Wen Deng
- Department of Biostatistics and Bioinformatics, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA, USA.
| | - Lei Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College of Soochow University, Jiangsu, PR China; Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College of Soochow University, Jiangsu, PR China.
| |
Collapse
|
43
|
Melguizo-Rodríguez L, Manzano-Moreno FJ, De Luna-Bertos E, Rivas A, Ramos-Torrecillas J, Ruiz C, García-Martínez O. Effect of olive oil phenolic compounds on osteoblast differentiation. Eur J Clin Invest 2018; 48. [PMID: 29392706 DOI: 10.1111/eci.12904] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 01/29/2018] [Indexed: 01/10/2023]
Abstract
BACKGROUND Osteoporosis is a skeletal disorder characterized by compromised bone strength that predisposes individuals to an increased risk of fracture. Previous in vivo and in vitro studies have reported that phenolic compounds present in extra virgin olive oil have a beneficial effect on osteoblasts in terms of increase cell proliferation. The aim of this study was to determine whether phenolic compounds present in olive oil could modify the expression of cell differentiation markers on osteoblasts. STUDY DESIGN An in vitro experimental design was performed using MG-63 osteoblasts cell line. METHODS MG63 cells were exposed to different doses of luteolin, apigenin, or p-coumaric, caffeic or ferulic acid. Alkaline phosphatase (ALP) was evaluated by spectrophotometry and antigen expression (cluster of differentiation [CD] 54, CD80, CD86 and HLA-DR) by flow cytometry. RESULTS At 24 hour, treated groups showed an increased ALP and modulated antigen profile, with respect to the nontreated group. CONCLUSION These results demonstrate that the phenolic compounds studied induce cell maturation in vitro, increasing ALP synthesis and reducing the expression of antigens involved in immune functions of the osteoblast which would improve bone density.
Collapse
Affiliation(s)
- Lucía Melguizo-Rodríguez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain
| | - Francisco Javier Manzano-Moreno
- Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain.,Biomedical Group (BIO277), Department of Stomatology, School of Dentistry, University of Granada, Granada, Spain
| | - Elvira De Luna-Bertos
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain
| | - Ana Rivas
- Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain.,AGR-255 Group, Department of Nutrition and Food Sciences, Faculty of Pharmacy, University of Granada, Granada, Spain
| | - Javier Ramos-Torrecillas
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain
| | - Concepción Ruiz
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain.,Institute of Neuroscience Federico Olóriz, University of Granada, Granada, Spain
| | - Olga García-Martínez
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences, University of Granada, Granada, Spain.,Instituto Investigación Biosanitaria, ibs.Granada, Granada, Spain
| |
Collapse
|
44
|
Manzano-Moreno FJ, Ramos-Torrecillas J, Melguizo-Rodríguez L, Illescas-Montes R, Ruiz C, García-Martínez O. Bisphosphonate Modulation of the Gene Expression of Different Markers Involved in Osteoblast Physiology: Possible Implications in Bisphosphonate-Related Osteonecrosis of the Jaw. Int J Med Sci 2018; 15:359-367. [PMID: 29511371 PMCID: PMC5835706 DOI: 10.7150/ijms.22627] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 01/05/2018] [Indexed: 12/28/2022] Open
Abstract
The aim of the present study was to elucidate the role of osteoblasts in bisphosphonates-related osteonecrosis of the jaw (BRONJ). The specific objective was to evaluate the effect on osteoblasts of two nitrogen-containing BPs (zoledronate and alendronate) and one non-nitrogen-containing BP (clodronate) by analyzing modulations in their expression of genes essential for osteoblast physiology. Real-time polymerase chain reaction (RT-PCR) was used to study the effects of zoledronate, alendronate, and clodronate at doses of 10-5, 10-7, or 10-9 M on the expression of Runx-2, OSX, ALP, OSC, OPG, RANKL, Col-I, BMP-2, BMP-7, TGF-β1, VEGF, TGF-βR1, TGF-βR2, and TGF-βR3 by primary human osteoblasts (HOBs) and MG-63 osteosarcoma cells. Expression of these markers was found to be dose-dependent, with no substantive differences between these cell lines. In general, results demonstrated a significant increase in TFG-β1, TGF-βR1, TGF-βR2, TGF-βR3, and VEGF expressions and a significant reduction in RUNX-2, Col-1, OSX, OSC, BMP-2, BMP-7, ALP, and RANKL expressions, while OPG expression varied according to the dose and cell line. The results of this in vitro study of HOBS and MG-63 cell lines indicate that low BP doses can significantly affect the expression of genes essential for osteoblast growth and differentiation and of genes involved in regulating osteoblast-osteoclast interaction, possibly by increasing TGF-β1 production. These findings suggest that osteoblasts may play an important role in BRONJ development, without ruling out other factors.
Collapse
Affiliation(s)
- Francisco Javier Manzano-Moreno
- Biomedical Group (BIO277), Department of Stomatology, School of Dentistry, University of Granada, Spain
- Instituto Investigación Biosanitaria, ibs.Granada (Spain)
| | - Javier Ramos-Torrecillas
- Instituto Investigación Biosanitaria, ibs.Granada (Spain)
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences. University of Granada, Spain
| | - Lucia Melguizo-Rodríguez
- Instituto Investigación Biosanitaria, ibs.Granada (Spain)
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences. University of Granada, Spain
| | - Rebeca Illescas-Montes
- Instituto Investigación Biosanitaria, ibs.Granada (Spain)
- Biomedical Group (BIO277), Department of Nursing, Faculty of Nursing, Melilla. University of Granada, Spain
| | - Concepción Ruiz
- Instituto Investigación Biosanitaria, ibs.Granada (Spain)
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences. University of Granada, Spain
- Institute of Neuroscience, Parque Tecnológico Ciencias de la Salud, Armilla (Granada), University of Granada, Spain
| | - Olga García-Martínez
- Instituto Investigación Biosanitaria, ibs.Granada (Spain)
- Biomedical Group (BIO277), Department of Nursing, Faculty of Health Sciences. University of Granada, Spain
| |
Collapse
|
45
|
Ring ES, Lawson MA, Snowden JA, Jolley I, Chantry AD. New agents in the Treatment of Myeloma Bone Disease. Calcif Tissue Int 2018; 102:196-209. [PMID: 29098361 PMCID: PMC5805798 DOI: 10.1007/s00223-017-0351-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Accepted: 10/19/2017] [Indexed: 12/17/2022]
Abstract
Patients with multiple myeloma develop a devastating bone disease driven by the uncoupling of bone remodelling, excess osteoclastic bone resorption and diminished osteoblastic bone formation. The bone phenotype is typified by focal osteolytic lesions leading to pathological fractures, hypercalcaemia and other catastrophic bone events such as spinal cord compression. This causes bone pain, impaired functional status, decreased quality of life and increased mortality. Early in the disease, malignant plasma cells occupy a niche environment that encompasses their interaction with other key cellular components of the bone marrow microenvironment. Through these interactions, osteoclast-activating factors and osteoblast inhibitory factors are produced, which together uncouple the dynamic process of bone remodelling, leading to net bone loss and focal osteolytic lesions. Current management includes antiresorptive therapies, i.e. bisphosphonates, palliative support and orthopaedic interventions. Bisphosphonates are the mainstay of treatment for myeloma bone disease (MBD), but are only partially effective and do have some significant disadvantages; for example, they do not lead to the repair of existing bone destruction. Thus, newer agents to prevent bone destruction and also promote bone formation and repair existing lesions are warranted. This review summarises novel ways that MBD is being therapeutically targeted.
Collapse
Affiliation(s)
- Elizabeth S Ring
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, The University of Sheffield Medical School, Beech Hill Road, Sheffield, South Yorkshire, S10 2RX, UK.
| | - Michelle A Lawson
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Mellanby Bone Centre, School of Medicine and Biomedical Sciences, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | - John A Snowden
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Ingrid Jolley
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, The University of Sheffield Medical School, Beech Hill Road, Sheffield, South Yorkshire, S10 2RX, UK
- Department of Radiology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| | - Andrew D Chantry
- Department of Oncology and Metabolism, Faculty of Medicine, Dentistry and Health, The University of Sheffield Medical School, Beech Hill Road, Sheffield, South Yorkshire, S10 2RX, UK
- Sheffield Myeloma Research Team, Department of Oncology and Metabolism, Mellanby Bone Centre, School of Medicine and Biomedical Sciences, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, UK
| |
Collapse
|
46
|
Hackinger S, Trajanoska K, Styrkarsdottir U, Zengini E, Steinberg J, Ritchie GRS, Hatzikotoulas K, Gilly A, Evangelou E, Kemp JP, Evans D, Ingvarsson T, Jonsson H, Thorsteinsdottir U, Stefansson K, McCaskie AW, Brooks RA, Wilkinson JM, Rivadeneira F, Zeggini E. Evaluation of shared genetic aetiology between osteoarthritis and bone mineral density identifies SMAD3 as a novel osteoarthritis risk locus. Hum Mol Genet 2018; 26:3850-3858. [PMID: 28934396 PMCID: PMC5886098 DOI: 10.1093/hmg/ddx285] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2017] [Accepted: 07/15/2017] [Indexed: 01/20/2023] Open
Abstract
Osteoarthritis (OA) is a common complex disease with high public health burden and no curative therapy. High bone mineral density (BMD) is associated with an increased risk of developing OA, suggesting a shared underlying biology. Here, we performed the first systematic overlap analysis of OA and BMD on a genome wide scale. We used summary statistics from the GEFOS consortium for lumbar spine (n = 31,800) and femoral neck (n = 32,961) BMD, and from the arcOGEN consortium for three OA phenotypes (hip, ncases=3,498; knee, ncases=3,266; hip and/or knee, ncases=7,410; ncontrols=11,009). Performing LD score regression we found a significant genetic correlation between the combined OA phenotype (hip and/or knee) and lumbar spine BMD (rg=0.18, P = 2.23 × 10−2), which may be driven by the presence of spinal osteophytes. We identified 143 variants with evidence for cross-phenotype association which we took forward for replication in independent large-scale OA datasets, and subsequent meta-analysis with arcOGEN for a total sample size of up to 23,425 cases and 236,814 controls. We found robustly replicating evidence for association with OA at rs12901071 (OR 1.08 95% CI 1.05–1.11, Pmeta=3.12 × 10−10), an intronic variant in the SMAD3 gene, which is known to play a role in bone remodeling and cartilage maintenance. We were able to confirm expression of SMAD3 in intact and degraded cartilage of the knee and hip. Our findings provide the first systematic evaluation of pleiotropy between OA and BMD, highlight genes with biological relevance to both traits, and establish a robust new OA genetic risk locus at SMAD3.
Collapse
Affiliation(s)
- Sophie Hackinger
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton CB10 1HH, UK
| | - Katerina Trajanoska
- Departments of Internal Medicine and Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | | | - Eleni Zengini
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK.,5th Department, Dromokaiteio Psychiatric Hospital, Athens 124 61, Greece
| | - Julia Steinberg
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton CB10 1HH, UK
| | | | | | - Arthur Gilly
- Human Genetics, Wellcome Trust Sanger Institute, Hinxton CB10 1HH, UK
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina 45110, Greece.,Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
| | - John P Kemp
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | | | - David Evans
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK.,University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, QLD, Australia
| | - Thorvaldur Ingvarsson
- Department of Orthopedic Surgery, Akureyri Hospital, 600 Akureyri, Iceland.,Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland.,Institution of Health Science, University of Akureyri, 600 Akureyri, Iceland
| | - Helgi Jonsson
- Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland.,Department of Medicine, Landspitali, The National University Hospital of Iceland, 101 Reykjavik, Iceland
| | - Unnur Thorsteinsdottir
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Kari Stefansson
- deCODE Genetics, Sturlugata 8, IS-101 Reykjavik, Iceland.,Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
| | - Andrew W McCaskie
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Box 180, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Roger A Brooks
- Division of Trauma & Orthopaedic Surgery, University of Cambridge, Box 180, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Jeremy M Wilkinson
- Department of Oncology and Metabolism, University of Sheffield, Sheffield S10 2RX, UK
| | - Fernando Rivadeneira
- Departments of Internal Medicine and Epidemiology, Erasmus University Medical Center, Rotterdam 3000 CA, The Netherlands
| | | |
Collapse
|
47
|
Hara Y, Ghazizadeh M, Shimizu H, Matsumoto H, Saito N, Yagi T, Mashiko K, Mashiko K, Kawai M, Yokota H. Delayed Expression of Circulating TGF-β1 and BMP-2 Levels in Human Nonunion Long Bone Fracture Healing. J NIPPON MED SCH 2017; 84:12-18. [PMID: 28331138 DOI: 10.1272/jnms.84.12] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The healing process of bone fracture requires a well-controlled multistage and sequential order beginning immediately after the injury. However, complications leading to nonunion exist, creating serious problems and costs for patients. Transforming growth factor-beta 1 (TGF-β1) and bone morphogenic protein 2 (BMP-2) are two major growth factors involved in human bone fracture healing by promoting various stages of bone ossification. In this study, we aimed to determine the role of these factors during the fracture healing of human long bones and assess their impacts on nonunion condition. MATERIALS AND METHODS We performed a comprehensive analysis of plasma TGF-β1 and BMP-2 levels in blood samples from 10 patients with proved nonunion and 10 matched patients with normal union following a predetermined time schedule. The concentrations of TGF-β1 and BMP-2 were measured at each time point using a solid-phase ELISA. RESULTS TGF-β1 and BMP-2 levels were detectable in all patients. For all patients, a maximal peak for TGF-β1 was found at 3-week. In normal union group, TGF-β1 showed a maximal peak at 2-week while nonunion group had a delayed maximal peak at 3-week. Plasma levels of BMP-2 for all patients and for normal union group reached a maximal peak at 1-week, but nonunion group showed a delayed maximal peak at 2-week. In general, plasma TGF-β1 or BMP-2 level was not significantly different between normal union and nonunion groups. CONCLUSION The expression levels of TGF-β1 and BMP-2 appeared to be delayed in nonunion patients which could play an important role in developing an early marker of fracture union condition and facilitate improved patient's management.
Collapse
Affiliation(s)
- Yoshiaki Hara
- Department of Emergency and Critical Care Medicine, Nippon Medical School Chiba Hokusoh Hospital
| | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Notsu M, Kanazawa I, Takeno A, Yokomoto-Umakoshi M, Tanaka KI, Yamaguchi T, Sugimoto T. Advanced Glycation End Product 3 (AGE3) Increases Apoptosis and the Expression of Sclerostin by Stimulating TGF-β Expression and Secretion in Osteocyte-Like MLO-Y4-A2 Cells. Calcif Tissue Int 2017; 100:402-411. [PMID: 28229177 DOI: 10.1007/s00223-017-0243-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 01/24/2017] [Indexed: 12/14/2022]
Abstract
Advanced glycation end products (AGEs) cause bone fragility due to deterioration in bone quality. We previously reported that AGE3 induced apoptosis and inhibited differentiation via increased transforming growth factor (TGF)-β signaling in osteoblastic cells. Additionally, we demonstrated that AGE3 increased apoptosis and sclerostin expression and decreased receptor activator of nuclear factor-κB ligand (RANKL) expression in osteocyte-like cells. However, it remains unclear whether TGF-β signaling is involved in the effects of AGEs on apoptosis and the expression of sclerostin and RANKL in osteocytes. Effects of AGE3 on apoptosis of mouse osteocyte-like MLO-Y4-A2 cells were examined by DNA fragmentation ELISA. Expression of TGF-β, sclerostin, and RANKL was evaluated using real-time PCR, Western blotting, and ELISA kits. To block TGF-β signaling, we used SD208, a TGF-β type I receptor kinase inhibitor. AGE3 (200 µg/mL) significantly increased apoptosis and mRNA expression of Sost, the gene encoding sclerostin, and decreased Rankl mRNA expression in MLO-Y4-A2 cells. AGE3 significantly increased the expression of TGF-β. Co-incubation of SD208 with AGE3 significantly rescued AGE3-induced apoptosis in a dose-dependent manner. Moreover, SD208 restored AGE3-increased mRNA and protein expression of sclerostin. In contrast, SD208 did not affect AGE3-decreased mRNA and protein expression of RANKL. These findings suggest that AGE3 increases apoptosis and sclerostin expression through increasing TGF-β expression in osteocytes, and that AGE3 decreases RANKL expression independent of TGF-β signaling.
Collapse
Affiliation(s)
- Masakazu Notsu
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Ippei Kanazawa
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan.
| | - Ayumu Takeno
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Maki Yokomoto-Umakoshi
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Ken-Ichiro Tanaka
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Toru Yamaguchi
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan
| | - Toshitsugu Sugimoto
- Internal Medicine 1, Shimane University Faculty of Medicine, 89-1, Enya-cho, Izumo, Shimane, 693-8501, Japan
| |
Collapse
|
49
|
Bi X, Grafe I, Ding H, Flores R, Munivez E, Jiang MM, Dawson B, Lee B, Ambrose CG. Correlations Between Bone Mechanical Properties and Bone Composition Parameters in Mouse Models of Dominant and Recessive Osteogenesis Imperfecta and the Response to Anti-TGF-β Treatment. J Bone Miner Res 2017; 32:347-359. [PMID: 27649409 PMCID: PMC7894383 DOI: 10.1002/jbmr.2997] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/12/2022]
Abstract
Osteogenesis imperfecta (OI) is a group of genetic disorders characterized by brittle bones that are prone to fracture. Although previous studies in animal models investigated the mechanical properties and material composition of OI bone, little work has been conducted to statistically correlate these parameters to identify key compositional contributors to the impaired bone mechanical behaviors in OI. Further, although increased TGF-β signaling has been demonstrated as a contributing mechanism to the bone pathology in OI models, the relationship between mechanical properties and bone composition after anti-TGF-β treatment in OI has not been studied. Here, we performed follow-up analyses of femurs collected in an earlier study from OI mice with and without anti-TGF-β treatment from both recessive (Crtap-/- ) and dominant (Col1a2+/P.G610C ) OI mouse models and WT mice. Mechanical properties were determined using three-point bending tests and evaluated for statistical correlation with molecular composition in bone tissue assessed by Raman spectroscopy. Statistical regression analysis was conducted to determine significant compositional determinants of mechanical integrity. Interestingly, we found differences in the relationships between bone composition and mechanical properties and in the response to anti-TGF-β treatment. Femurs of both OI models exhibited increased brittleness, which was associated with reduced collagen content and carbonate substitution. In the Col1a2+/P.G610C femurs, reduced hydroxyapatite crystallinity was also found to be associated with increased brittleness, and increased mineral-to-collagen ratio was correlated with increased ultimate strength, elastic modulus, and bone brittleness. In both models of OI, regression analysis demonstrated that collagen content was an important predictor of the increased brittleness. In summary, this work provides new insights into the relationships between bone composition and material properties in models of OI, identifies key bone compositional parameters that correlate with the impaired mechanical integrity of OI bone, and explores the effects of anti-TGF-β treatment on bone-quality parameters in these models. © 2016 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Xiaohong Bi
- Department of Nanomedicine and Biomedical Engineering, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Ingo Grafe
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Hao Ding
- Department of Nanomedicine and Biomedical Engineering, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rene Flores
- Academic and Research Affairs, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Elda Munivez
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ming Ming Jiang
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brian Dawson
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Brendan Lee
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Catherine G Ambrose
- Department of Orthopaedic Surgery, University of Texas Health Science Center at Houston, Houston, TX, USA
| |
Collapse
|
50
|
Ripamonti U, Parak R, Klar RM, Dickens C, Dix-Peek T, Duarte R. Cementogenesis and osteogenesis in periodontal tissue regeneration by recombinant human transforming growth factor-β3: a pilot studyin Papio ursinus. J Clin Periodontol 2016; 44:83-95. [DOI: 10.1111/jcpe.12642] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/22/2016] [Indexed: 12/26/2022]
Affiliation(s)
- Ugo Ripamonti
- Bone Research Laboratory; Department of Oral Medicine & Periodontology; School of Oral Health Sciences; Faculty of Health Sciences; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
| | - Ruqayya Parak
- Bone Research Laboratory; Department of Oral Medicine & Periodontology; School of Oral Health Sciences; Faculty of Health Sciences; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
- Department of Oral Biological Sciences; Faculty of Health Sciences; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
| | - Roland M. Klar
- Bone Research Laboratory; Department of Oral Medicine & Periodontology; School of Oral Health Sciences; Faculty of Health Sciences; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
- Department of Internal Medicine; Faculty of Health Sciences; School of Clinical Medicine; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
| | - Caroline Dickens
- Department of Internal Medicine; Faculty of Health Sciences; School of Clinical Medicine; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
| | - Therese Dix-Peek
- Department of Internal Medicine; Faculty of Health Sciences; School of Clinical Medicine; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
| | - Raquel Duarte
- Department of Internal Medicine; Faculty of Health Sciences; School of Clinical Medicine; University of the Witwatersrand, Johannesburg; Johannesburg South Africa
| |
Collapse
|