1
|
Zhang X, Tian L, Majumdar A, Scheller EL. Function and Regulation of Bone Marrow Adipose Tissue in Health and Disease: State of the Field and Clinical Considerations. Compr Physiol 2024; 14:5521-5579. [PMID: 39109972 PMCID: PMC11725182 DOI: 10.1002/cphy.c230016] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2025]
Abstract
Bone marrow adipose tissue (BMAT) is a metabolically and clinically relevant fat depot that exists within bone. Two subtypes of BMAT, regulated and constitutive, reside in hematopoietic-rich red marrow and fatty yellow marrow, respectively, and exhibit distinct characteristics compared to peripheral fat such as white and brown adipose tissues. Bone marrow adipocytes (BMAds) are evolutionally preserved in most vertebrates, start development after birth and expand throughout life, and originate from unique progenitor populations that control bone formation and hematopoiesis. Mature BMAds also interact closely with other cellular components of the bone marrow niche, serving as a nearby energy reservoir to support the skeletal system, a signaling hub that contributes to both local and systemic homeostasis, and a final fuel reserve for survival during starvation. Though BMAT and bone are often inversely correlated, more BMAT does not always mean less bone, and the prevention of BMAT expansion as a strategy to prevent bone loss remains questionable. BMAT adipogenesis and lipid metabolism are regulated by the nervous systems and a variety of circulating hormones. This contributes to the plasticity of BMAT, including BMAT expansion in common physiological or pathological conditions, and BMAT catabolism under certain extreme circumstances, which are often associated with malnutrition and/or systemic inflammation. Altogether, this article provides a comprehensive overview of the local and systemic functions of BMAT and discusses the regulation and plasticity of this unique adipose tissue depot in health and disease. © 2024 American Physiological Society. Compr Physiol 14:5521-5579, 2024.
Collapse
Affiliation(s)
- Xiao Zhang
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Linda Tian
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
| | - Anurag Majumdar
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
| | - Erica L. Scheller
- Division of Bone and Mineral Diseases, Department of Medicine, Washington University, St. Louis, Missouri, USA
- Department of Biomedical Engineering, Washington University, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University, St. Louis, Missouri, USA
| |
Collapse
|
2
|
Gómez R, Barter MJ, Alonso-Pérez A, Skelton AJ, Proctor C, Herrero-Beaumont G, Young DA. DNA methylation analysis identifies key transcription factors involved in mesenchymal stem cell osteogenic differentiation. Biol Res 2023; 56:9. [PMID: 36890579 PMCID: PMC9996951 DOI: 10.1186/s40659-023-00417-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 01/23/2023] [Indexed: 03/10/2023] Open
Abstract
BACKGROUND Knowledge about regulating transcription factors (TFs) for osteoblastogenesis from mesenchymal stem cells (MSCs) is limited. Therefore, we investigated the relationship between genomic regions subject to DNA-methylation changes during osteoblastogenesis and the TFs known to directly interact with these regulatory regions. RESULTS The genome-wide DNA-methylation signature of MSCs differentiated to osteoblasts and adipocytes was determined using the Illumina HumanMethylation450 BeadChip array. During adipogenesis no CpGs passed our test for significant methylation changes. Oppositely, during osteoblastogenesis we identified 2462 differently significantly methylated CpGs (adj. p < 0.05). These resided outside of CpGs islands and were significantly enriched in enhancer regions. We confirmed the correlation between DNA-methylation and gene expression. Accordingly, we developed a bioinformatic tool to analyse differentially methylated regions and the TFs interacting with them. By overlaying our osteoblastogenesis differentially methylated regions with ENCODE TF ChIP-seq data we obtained a set of candidate TFs associated to DNA-methylation changes. Among them, ZEB1 TF was highly related with DNA-methylation. Using RNA interference, we confirmed that ZEB1, and ZEB2, played a key role in adipogenesis and osteoblastogenesis processes. For clinical relevance, ZEB1 mRNA expression in human bone samples was evaluated. This expression positively correlated with weight, body mass index, and PPARγ expression. CONCLUSIONS In this work we describe an osteoblastogenesis-associated DNA-methylation profile and, using these data, validate a novel computational tool to identify key TFs associated to age-related disease processes. By means of this tool we identified and confirmed ZEB TFs as mediators involved in the MSCs differentiation to osteoblasts and adipocytes, and obesity-related bone adiposity.
Collapse
Affiliation(s)
- Rodolfo Gómez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Laboratorio 18, Edificio B, Planta -2, 15706, Santiago de Compostela, Spain.
| | - Matt J Barter
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE1 3BZ, UK
| | - Ana Alonso-Pérez
- Musculoskeletal Pathology Group, Institute IDIS, Santiago University Clinical Hospital, Laboratorio 18, Edificio B, Planta -2, 15706, Santiago de Compostela, Spain
| | - Andrew J Skelton
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE1 3BZ, UK
- Bioinformatics Support Unit, Faculty of Medical Sciences, Newcastle University, Newcastle-upon-Tyne, NE2 4HH, UK
| | - Carole Proctor
- Campus for Ageing and Vitality, Newcastle University, Newcastle-Upon-Tyne, NE2 4HH, UK
| | - Gabriel Herrero-Beaumont
- Bone and Joint Research Unit, IIS-Fundación Jiménez Díaz, UAM, 28040, Madrid, Avda Reyes Católicos, Spain
| | - David A Young
- Skeletal Research Group, Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, NE1 3BZ, UK
| |
Collapse
|
3
|
Mehta D, Dankert J, Yim N, Leclerc K, Leucht P. Rosiglitazone induces adipogenesis of both marrow and periosteum derived mesenchymal stem cells during endochondral fracture healing. J Orthop Sci 2023; 28:460-467. [PMID: 34879982 PMCID: PMC9167886 DOI: 10.1016/j.jos.2021.11.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Revised: 10/21/2021] [Accepted: 11/10/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Type 2 diabetes mellitus (T2DM) afflicts about six percent of the global population, and these patients suffer from a two-fold increased fracture risk. Thiazolidinediones (TZDs), including rosiglitazone, are commonly used medications in T2DM because they have a low incidence of monotherapy failure. It is known that rosiglitazone is associated with secondary osteoporosis, further increasing the fracture risk in an already susceptible population. However, it is not yet understood how rosiglitazone impacts endochondral bone healing after fracture. The aim of this study is to elucidate how rosiglitazone treatment impacts endochondral fracture healing, and how rosiglitazone influences the differentiation of skeletal stem and progenitor cells from the bone marrow and the periosteum. METHODS An in-vivo mouse femur fracture model was employed to evaluate differences in fracture healing between mice treated with and without rosiglitazone chow. Fracture healing was assessed with histology and micro computed tomography (μCT). In-vitro assays utilized isolated mouse bone marrow stromal cells and periosteal cells to investigate how rosiglitazone impacts the osteogenic capability and adipogenicity of these cells. RESULTS The in-vivo mouse femur fracture model showed that fracture callus in mice treated with rosiglitazone had significantly more adipose content than those of control mice that did not receive rosiglitazone. In addition, μCT analysis showed that rosiglitazone treated mice had significantly greater bone volume, but overall greater porosity when compared to control mice. In-vitro experimentation showed significantly less osteogenesis and more adipogenesis in bone marrow derived progenitor cells that were cultured in osteogenic media. In addition, rosiglitazone treatment alone caused significant increases in adipogenesis in both bone marrow and periosteum derived cells. CONCLUSION Rosiglitazone impairs endochondral fracture healing in mice by increasing adipogenesis and decreasing osteogenesis of both bone marrow and periosteum derived skeletal progenitor cells.
Collapse
Affiliation(s)
- Devan Mehta
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - John Dankert
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - Nury Yim
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - Kevin Leclerc
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| | - Philipp Leucht
- NYU Grossman School of Medicine - NYU Langone Orthopedic Hospital, Department of Orthopedic Surgery, New York, NY, USA.
| |
Collapse
|
4
|
Chougule A, Baroi S, Czernik PJ, Crowe E, Chang MR, Griffin PR, Lecka-Czernik B. Osteocytes contribute via nuclear receptor PPAR-alpha to maintenance of bone and systemic energy metabolism. Front Endocrinol (Lausanne) 2023; 14:1145467. [PMID: 37181042 PMCID: PMC10173151 DOI: 10.3389/fendo.2023.1145467] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2023] [Accepted: 03/20/2023] [Indexed: 05/16/2023] Open
Abstract
Introduction The view that bone and energy metabolism are integrated by common regulatory mechanisms is broadly accepted and supported by multiple strands of evidence. This includes the well-characterized role of the PPARγ nuclear receptor, which is a common denominator in energy metabolism and bone metabolism. Little is known, however, about the role of PPARα nuclear receptor, a major regulator of lipid metabolism in other organs, in bone. Methods A side-by-side comparative study of 5-15 mo old mice with global PPARα deficiency (αKO) and mice with osteocyte-specific PPARα deficiency (αOTKO) in order to parse out the various activities of PPARα in the skeleton that are of local and systemic significance. This study included transcriptome analysis of PPARα-deficient osteocytes, and analyses of bone mass and bone microarchitecture, systemic energy metabolism with indirect calorimetry, and differentiation potential of hematopoietic and mesenchymal bone cell progenitors. These analyses were paired with in vitro studies of either intact or silenced for PPARα MLO-A5 cells to determine PPARα role in osteocyte bioenergetics. Results In osteocytes, PPARα controls large number of transcripts coding for signaling and secreted proteins which may regulate bone microenvironment and peripheral fat metabolism. In addition, PPARα in osteocytes controls their bioenergetics and mitochondrial response to stress, which constitutes up to 40% of total PPARα contribution to the global energy metabolism. Similarly to αKO mice, the metabolic phenotype of αOTKO mice (both males and females) is age-dependent. In younger mice, osteocyte metabolism contributes positively to global energetics, however, with aging the high-energy phenotype reverts to a low-energy phenotype and obesity develops, suggesting a longitudinal negative effect of impaired lipid metabolism and mitochondrial dysfunction in osteocytes deficient in PPARα. However, bone phenotype was not affected in αOTKO mice except in the form of an increased volume of marrow adipose tissue in males. In contrast, global PPARα deficiency in αKO mice led to enlarged bone diameter with a proportional increase in number of trabeculae and enlarged marrow cavities; it also altered differentiation of hematopoietic and mesenchymal marrow cells toward osteoclast, osteoblast and adipocyte lineages, respectively. Discussion PPARα role in bone is multileveled and complex. In osteocytes, PPARα controls the bioenergetics of these cells, which significantly contributes to systemic energy metabolism and their endocrine/paracrine function in controlling marrow adiposity and peripheral fat metabolism.
Collapse
Affiliation(s)
- Amit Chougule
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Sudipta Baroi
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Piotr J. Czernik
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Emily Crowe
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
| | - Mi Ra Chang
- Department of Molecular Medicine, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, United States
| | - Patrick R. Griffin
- Department of Molecular Medicine, UF Scripps Biomedical Research, University of Florida, Jupiter, FL, United States
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- Department of Physiology and Pharmacology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH, United States
- *Correspondence: Beata Lecka-Czernik,
| |
Collapse
|
5
|
Tanios M, Brickman B, Cage E, Abbas K, Smith C, Atallah M, Baroi S, Lecka-Czernik B. Diabetes and Impaired Fracture Healing: A Narrative Review of Recent Literature. Curr Osteoporos Rep 2022; 20:229-239. [PMID: 35960475 DOI: 10.1007/s11914-022-00740-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/22/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF THE REVIEW Diabetes mellitus is a chronic metabolic disorder commonly encountered in orthopedic patients. Both type 1 and type 2 diabetes mellitus increase fracture risk and impair fracture healing. This review examines complex etiology of impaired fracture healing in diabetes. RECENT FINDINGS Recent findings point to several mechanisms leading to orthopedic complications in diabetes. Hyperglycemia and chronic inflammation lead to increased formation of advanced glycation end products and generation of reactive oxygen species, which in turn contribute to the disruption in osteoblast and osteoclast balance leading to decreased bone formation and heightening the risk of nonunion or delayed union as well as impaired fracture healing. The mechanisms attributing to this imbalance is secondary to an increase in pro-inflammatory mediators leading to premature resorption of callus cartilage and impaired bone formation due to compromised osteoblast differentiation and their apoptosis. Other mechanisms include disruption in the bone's microenvironment supporting different stages of healing process including hematoma and callus formation, and their resolution during bone remodeling phase. Complications of diabetes including peripheral neuropathy and peripheral vascular disease also contribute to the impairment of fracture healing. Certain diabetic drugs may have adverse effects on fracture healing. The pathophysiology of impaired fracture healing in diabetic patients is complex. This review provides an update of the most recent findings on how key mediators of bone healing are affected in diabetes.
Collapse
Affiliation(s)
- Mina Tanios
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| | - Bradley Brickman
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Emily Cage
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Kassem Abbas
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Cody Smith
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Marina Atallah
- The University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Sudipta Baroi
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA
| | - Beata Lecka-Czernik
- Department of Orthopedic Surgery, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine and Life Sciences, Toledo, OH, USA.
| |
Collapse
|
6
|
Boregowda SV, Nanjappa MK, Booker CN, Strivelli J, Supper VM, Cooke PS, Phinney DG. Pharmacological Inhibition of Inositol Hexakisphosphate Kinase 1 Protects Mice against Obesity-Induced Bone Loss. BIOLOGY 2022; 11:biology11091257. [PMID: 36138736 PMCID: PMC9495776 DOI: 10.3390/biology11091257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 08/17/2022] [Accepted: 08/18/2022] [Indexed: 11/16/2022]
Abstract
Obesity and type II diabetes mellitus (T2DM) are prominent risk factors for secondary osteoporosis due to the negative impacts of hyperglycemia and excessive body fat on bone metabolism. While the armamentarium of anti-diabetic drugs is expanding, their negative or unknown impacts on bone metabolism limits effectiveness. The inactivation of inositol hexakisphosphate kinase 1 (IP6K1) protects mice from high-fat-diet (HFD)-induced obesity (DIO) and insulin resistance by enhancing thermogenic energy expenditure, but the role of this kinase and the consequences of its inhibition on bone metabolism are unknown. To determine if IP6K1 inhibition in obese mice affords protection against obesity-induced metabolic derangements and bone loss, we maintained 2-month-old mice on a normal chow control diet or HFD under thermal neutral conditions for 100 d. Beginning on day 40, HFD-fed mice were divided into two groups and administered daily injections of vehicle or the pan-IP6K inhibitor TNP [N2-(m-Trifluorobenzyl), N6-(p-nitrobenzyl) purine]. HFD-fed mice developed obesity, hyperglycemia, hyperlipidemia, and secondary osteoporosis, while TNP administration protected mice against HFD-induced metabolic and lipid derangements and preserved bone mass, mineral density, and trabecular microarchitecture, which correlated with reduced serum leptin levels, reduced marrow adiposity, and preservation of marrow resident skeletal stem/progenitor cells (SSPCs). TNP also exhibited hypotensive activity, an unrealized benefit of the drug, and its prolonged administration had no adverse impacts on spermatogenesis. Together, these data indicate that the inhibition of IP6K1 using selective inhibitors, such as TNP, may provide an effective strategy to manage obesity and T2DM due to its bone sparing effects.
Collapse
Affiliation(s)
- Siddaraju V. Boregowda
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | | | - Cori N. Booker
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Jacqueline Strivelli
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
| | - Valentina M. Supper
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32610, USA
| | - Paul S. Cooke
- Department of Physiological Sciences, University of Florida, Gainesville, FL 32610, USA
| | - Donald G. Phinney
- Department of Molecular Medicine, UF Scripps Biomedical Research, Jupiter, FL 33458, USA
- Correspondence:
| |
Collapse
|
7
|
Gaculenko A, Gregoric G, Popp V, Seyler L, Ringer M, Kachler K, Wu Z, Kisel W, Hofbauer C, Hofbauer LC, Uder M, Schett G, Bäuerle T, Bozec A. Systemic PPARγ Antagonism Reduces Metastatic Tumor Progression in Adipocyte-Rich Bone in Excess Weight Male Rodents. J Bone Miner Res 2021; 36:2440-2452. [PMID: 34378824 DOI: 10.1002/jbmr.4422] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022]
Abstract
Primary tumors are widely associated with an excess in body fat. The role of adipose tissue on tumor cell homing to bone is yet poorly defined. In this study, we aimed to assess whether bone colonization by tumor cells is favored by an adipocyte-rich bone marrow. We delineated the accompanying alterations of the bone microenvironment and established a treatment approach that interferes with high fat diet (HFD)-induced bone metastasis formation. We were able to show that adipocytes affect skeletal tumor growth in a metastatic model of breast cancer in male rats and melanoma in male mice as well as in human breast cancer bone biopsies. Indeed, HFD-induced bone marrow adiposity was accompanied by accelerated tumor progression and increased osteolytic lesions. In human bone metastases, bone marrow adiposity correlated with tumor cell proliferation. By antagonization of the adipocyte differentiation and storage pathway linked to the peroxisome proliferator-activated receptor gamma (PPARγ) with bisphenol-A-diglycidylether (BADGE), we were able to decelerate tumor progression and subsequent osteolytic damage in the bones of two distinct metastatic animal models exposed to HFD. Overall these data show that adipose tissue is a critical factor in bone metastases and cancer-induced bone loss. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Anastasia Gaculenko
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Institute of Radiology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Gasper Gregoric
- Institute of Radiology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Vanessa Popp
- Institute of Radiology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Lisa Seyler
- Institute of Radiology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Mark Ringer
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katerina Kachler
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Zhengquan Wu
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wadim Kisel
- University Center for Traumatology, Orthopedics and Plastic Surgery OUPC, Technische Universität Dresden, Dresden, Germany
| | - Christine Hofbauer
- National Center for Tumor Diseases (NCT), Partner Site Dresden/University Cancer Center (UCC), Technische Universität Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III and University Center for Healthy Aging, Technische Universität Dresden, Dresden, Germany
| | - Michael Uder
- Institute of Radiology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Georg Schett
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Tobias Bäuerle
- Institute of Radiology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Aline Bozec
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich Alexander University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| |
Collapse
|
8
|
Mucke HAM. Drug Repurposing Patent Applications July-September 2020. Assay Drug Dev Technol 2021; 19:204-208. [PMID: 33606552 DOI: 10.1089/adt.2020.1072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
9
|
Chougule A, Kolli V, Baroi S, Ebraheim N, Czernik PJ, Loh YP, Lecka-Czernik B. Nonenzymatic and Trophic Activities of Carboxypeptidase E Regulate Bone Mass and Bioenergetics of Skeletal Stem Cells in Mice. JBMR Plus 2020; 4:e10392. [PMID: 32995694 PMCID: PMC7507073 DOI: 10.1002/jbm4.10392] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/24/2020] [Accepted: 06/30/2020] [Indexed: 12/20/2022] Open
Abstract
Bone and energy metabolism are integrated by common regulatory mechanisms. Carboxypeptidase E (CPE), also known as obesity susceptibility protein or neurotrophic factor‐α1, is recognized for its function in processing prohormones, including proinsulin and pro‐opiomelanocortin polypeptide. Independent of its enzymatic activity, CPE may also act as a secreted factor with divergent roles in neuroprotection and cancer growth; however, its role in the regulation of bone mass and skeletal cell differentiation is unknown. Male mice with global deficiency in CPE are characterized with profound visceral obesity, low bone mass in both appendicular and axial skeleton, and high volume of marrow fat. Interestingly, although metabolic deficit of CPE KO mice develops early in life, bone deficit develops in older age, suggesting that CPE bone‐specific activities differ from its enzymatic activities. Indeed, mutated CPE knockin (mCPE KI) mice ectopically expressing CPE‐E342Q, a mutated protein lacking enzymatic activity, develop the same obese phenotype and accumulate the same volume of marrow fat as CPE KO mice, but their bone mass is normal. In addition, differentiation of marrow hematopoietic cells toward tartrate‐resistant acid phosphatase‐positive multinucleated osteoclasts is highly increased in CPE KO mice, but normal in mCPE KI mice. Moreover, in murine skeletal stem cells, nonenzymatic trophic CPE has activated ERK signaling, increased cell proliferation and increased mitochondrial activity. Treatment of preosteoblastic cells with intact or mutated recombinant CPE led to a transient accumulation of small lipid droplets, increased oxidative phosphorylation, and increased cellular dependence on fatty acids as fuel for energy production. In human marrow aspirates, CPE expression increases up to 30‐fold in osteogenic conditions. These findings suggest that nonenzymatic and trophic activities of CPE regulate bone mass, whereas marrow adiposity is controlled by CPE enzymatic activity. Thus, CPE can be positioned as a factor regulating simultaneously bone and energy metabolism through a combination of shared and distinct mechanisms. © 2020 The Authors. JBMR Plus published by Wiley Periodicals, Inc. on behalf of American Society for Bone and Mineral Research © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Amit Chougule
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Center for Diabetes and Endocrine Research University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Vipula Kolli
- Section on Cellular Neurobiology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health Bethesda MD USA
| | - Sudipta Baroi
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Center for Diabetes and Endocrine Research University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Nabil Ebraheim
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Piotr J Czernik
- Department of Physiology and Pharmacology University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| | - Y Peng Loh
- Section on Cellular Neurobiology Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health Bethesda MD USA
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Department of Physiology and Pharmacology University of Toledo, College of Medicine and Life Sciences Toledo OH USA.,Center for Diabetes and Endocrine Research University of Toledo, College of Medicine and Life Sciences Toledo OH USA
| |
Collapse
|
10
|
Xu H, Yang W, Chen P, Chen R, Xue P, Wang L, Yuan J, Yao Q, Chen B, Zhao Y. Bone‐Inspired Tube Filling Decellularized Matrix of Toad Cartilage Provided an Osteoinductive Microenvironment for Mesenchymal Stem Cells to Facilitate the Radius Defect Repair of Rabbit. Biotechnol J 2020; 15:e2000004. [DOI: 10.1002/biot.202000004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 04/18/2020] [Indexed: 12/13/2022]
Affiliation(s)
- He‐Lin Xu
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Wai‐Geng Yang
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Pian‐Pian Chen
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Rui Chen
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Peng‐Peng Xue
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Li‐Fen Wang
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Jian‐Dong Yuan
- Department of OrthopedicsThe First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang 325000 P. R. China
| | - Qing Yao
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| | - Bin Chen
- Department of UltrasonographyThe First Affiliated Hospital of Wenzhou Medical University Wenzhou Zhejiang Province 325000 China
| | - Ying‐Zheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical SciencesWenzhou Medical University Wenzhou Zhejiang Province 325035 China
| |
Collapse
|
11
|
Pagnotti GM, Styner M, Uzer G, Patel VS, Wright LE, Ness KK, Guise TA, Rubin J, Rubin CT. Combating osteoporosis and obesity with exercise: leveraging cell mechanosensitivity. Nat Rev Endocrinol 2019; 15:339-355. [PMID: 30814687 PMCID: PMC6520125 DOI: 10.1038/s41574-019-0170-1] [Citation(s) in RCA: 147] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Osteoporosis, a condition of skeletal decline that undermines quality of life, is treated with pharmacological interventions that are associated with poor adherence and adverse effects. Complicating efforts to improve clinical outcomes, the incidence of obesity is increasing, predisposing the population to a range of musculoskeletal complications and metabolic disorders. Pharmacological management of obesity has yet to deliver notable reductions in weight and debilitating complications are rarely avoided. By contrast, exercise shows promise as a non-invasive and non-pharmacological method of regulating both osteoporosis and obesity. The principal components of exercise - mechanical signals - promote bone and muscle anabolism while limiting formation and expansion of fat mass. Mechanical regulation of bone and marrow fat might be achieved by regulating functions of differentiated cells in the skeletal tissue while biasing lineage selection of their common progenitors - mesenchymal stem cells. An inverse relationship between adipocyte versus osteoblast fate selection from stem cells is implicated in clinical conditions such as childhood obesity and increased marrow adiposity in type 2 diabetes mellitus, as well as contributing to skeletal frailty. Understanding how exercise-induced mechanical signals can be used to improve bone quality while decreasing fat mass and metabolic dysfunction should lead to new strategies to treat chronic diseases such as osteoporosis and obesity.
Collapse
Affiliation(s)
- Gabriel M Pagnotti
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Maya Styner
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Gunes Uzer
- College of Mechanical and Biomedical Engineering, Boise State University, Boise, ID, USA
| | - Vihitaben S Patel
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Laura E Wright
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Kirsten K Ness
- Department of Epidemiology and Cancer Control, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Theresa A Guise
- School of Medicine, Division of Endocrinology, Indiana University, Indianapolis, IN, USA
| | - Janet Rubin
- Department of Medicine, Division of Endocrinology and Metabolism, University of North Carolina, Chapel Hill, NC, USA
| | - Clinton T Rubin
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA.
| |
Collapse
|
12
|
Exploring conditions that make cortical bone geometry optimal for physiological loading. Biomech Model Mechanobiol 2019; 18:1335-1349. [PMID: 30953214 DOI: 10.1007/s10237-019-01147-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Accepted: 04/02/2019] [Indexed: 10/27/2022]
Abstract
While physiological loading on lower long bones changes during bone development, the bone cross section either remains circular or slowly changes from nearly circular to other shapes such as oval and roughly triangular. Bone is said to be an optimal structure, where strength is maximized using the optimal distribution of bone mass (also called Wolff's law). One of the most appropriate mathematical validations of this law would be a structural optimization-based formulation where total strain energy is minimized against a mass and a space constraint. Assuming that the change in cross section during bone development and homeostasis after adulthood is direct result of the change in physiological loading, this work investigates what optimization problem formulation (collectively, design variables, objective function, constraints, loading conditions, etc.) results in mathematically optimal solutions that resemble bones under actual physiological loading. For this purpose, an advanced structural optimization-based computational model for cortical bone development and defect repair is presented. In the optimization problem, overall bone stiffness is maximized first against a mass constraint, and then also against a polar first moment of area constraint that simultaneously constrains both mass and space. The investigation is completed in two stages. The first stage is developmental stage when physiological loading on lower long bones (tibia) is a random combination of axial, bending and torsion. The topology optimization applied to this case with the area moment constraint results into circular and elliptical cross sections similar to that found in growing mouse or human. The second investigation stage is bone homeostasis reached in adulthood when the physiological loading has a fixed pattern. A drill hole defect is applied to the adult mouse bone, which would disrupt the homeostasis. The optimization applied after the defect interestingly brings the damaged section back to the original intact geometry. The results, however, show that cortical bone geometry is optimal for the physiological loading only when there is also a constraint on polar moment of area. Further numerical experiments show that application of torsion along with the gait-analysis-based physiological loading improves the results, which seems to indicate that the cortical bone geometry is optimal for some amount of torsion in addition to the gait-based physiological loading. This work has a potential to be extended to bone growth/development models and fracture healing models, where topology optimization and polar moment of area constraint have not been introduced earlier.
Collapse
|
13
|
Li Y, Meng Y, Yu X. The Unique Metabolic Characteristics of Bone Marrow Adipose Tissue. Front Endocrinol (Lausanne) 2019; 10:69. [PMID: 30800100 PMCID: PMC6375842 DOI: 10.3389/fendo.2019.00069] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 01/24/2019] [Indexed: 02/05/2023] Open
Abstract
Bone marrow adipose tissue (MAT) is distinct from white adipose tissue (WAT) or brown adipose tissue (BAT) for its location, feature and function. As a largely ignored adipose depot, it is situated in bone marrow space and resided with bone tissue side-by-side. MAT is considered not only as a regulator of bone metabolism through paracrine, but also as a functionally particular adipose tissue that may contribute to global metabolism. Adipokines, inflammatory factors and other molecules derived from bone marrow adipocytes may exert systematic effects. In this review, we summary the evidence from several aspects including development, distribution, histological features and phenotype to elaborate the basic characteristics of MAT. We discuss the association between bone metabolism and MAT, and highlight our current understanding of this special adipose tissue. We further demonstrate the probable relationship between MAT and energy metabolism, as well as glucose metabolism. On the basis of preliminary results from animal model and clinical studies, we propose that MAT has its unique secretory and metabolic function, although there is no in-depth study at present.
Collapse
Affiliation(s)
- Yujue Li
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yang Meng
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Orthopedics, West China Hospital, Sichuan University, Chengdu, China
| | - Xijie Yu
- Laboratory of Endocrinology and Metabolism, Department of Endocrinology and Metabolism and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- *Correspondence: Xijie Yu ;
| |
Collapse
|
14
|
Berberine derivative, Q8, stimulates osteogenic differentiation. Biochem Biophys Res Commun 2018; 504:340-345. [PMID: 30190123 DOI: 10.1016/j.bbrc.2018.08.192] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Accepted: 08/29/2018] [Indexed: 12/22/2022]
Abstract
Berberine has been implicated to be involved in maintaining bone health due to its anti-oxidative and osteogenic properties. However, low potency and low bioavailability limit the clinical development of the drug. To overcome these obstacles, we previously synthesized a compound, Q8, which is a structural homolog of berberine. The present study examined the pharmacological functions of Q8 to evaluate its potential use in bone regeneration with respect to osteoblast differentiation. Here, we report that Q8 enhanced BMP4-induced alkaline phosphatase (ALP) activity and transcription from the ALP promoter. In addition, Q8 suppressed the expression and activity of PPARγ (a known negative regulator of osteogenesis due to its stimulatory effects on adipogenesis and its role as an adipogenic transcription factor), which in turn increases β-catenin expression in the nucleus, and ultimately promotes osteoblast differentiation. Meanwhile, Q8 reversed the inhibitory effects of the PPARγ agonist, rosiglitazone, on osteoblast differentiation. This study demonstrated that Q8 promotes osteoblast differentiation via inhibition of PPARγ and the enhancement of osteoblast function by Q8 may contribute to the prevention for osteoporosis.
Collapse
|
15
|
Paschou SA, Dede AD, Anagnostis PG, Vryonidou A, Morganstein D, Goulis DG. Type 2 Diabetes and Osteoporosis: A Guide to Optimal Management. J Clin Endocrinol Metab 2017; 102:3621-3634. [PMID: 28938433 DOI: 10.1210/jc.2017-00042] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Accepted: 05/18/2017] [Indexed: 12/15/2022]
Abstract
CONTEXT Both type 2 diabetes (T2D) and osteoporosis are affected by aging and quite often coexist. Furthermore, the fracture risk in patients with T2D is increased. The aim of this article is to review updated information on osteoporosis and fracture risk in patients with T2D, to discuss the effects of diabetes treatment on bone metabolism, as well as the effect of antiosteoporotic medications on the incidence and control of T2D, and to provide a personalized guide to the optimal management. EVIDENCE ACQUISITION A systematic literature search for human studies was conducted in three electronic databases (PubMed, Cochrane, and EMBASE) until March 2017. Regarding recommendations, we adopted the grading system introduced by the American College of Physicians. EVIDENCE SYNTHESIS The results are presented in systematic tables. Healthy diet and physical exercise are very important for the prevention and treatment of both entities. Metformin, sulfonylureas, dipeptidyl peptidase-4 inhibitors, and glucagon-like peptide-1 receptor agonists should be preferred for the treatment of T2D in these patients, whereas strict targets should be avoided for the fear of hypoglycemia, falls, and fractures. Insulin should be used with caution and with careful measures to avoid hypoglycemia. Thiazolidinediones and canagliflozin should be avoided, whereas other sodium-dependent glucose transporter 2 inhibitors are less well-validated options. Insulin therapy is the preferred method for achieving glycemic control in hospitalized patients with T2D and fractures. The treatment and monitoring of osteoporosis should be continued without important amendments because of the presence of T2D. CONCLUSIONS Patients with coexisting T2D and osteoporosis should be managed in an optimal way according to scientific evidence.
Collapse
Affiliation(s)
- Stavroula A Paschou
- Division of Endocrinology and Diabetes, Aghia Sophia Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Anastasia D Dede
- Department of Endocrinology and Diabetes, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Panagiotis G Anagnostis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Andromachi Vryonidou
- Department of Endocrinology and Diabetes, Hellenic Red Cross Hospital, 11526 Athens, Greece
| | - Daniel Morganstein
- Department of Endocrinology and Diabetes, Chelsea and Westminster Hospital, London SW10 9NH, United Kingdom
| | - Dimitrios G Goulis
- Unit of Reproductive Endocrinology, First Department of Obstetrics and Gynecology, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
16
|
Davidson MA, Mattison DR, Azoulay L, Krewski D. Thiazolidinedione drugs in the treatment of type 2 diabetes mellitus: past, present and future. Crit Rev Toxicol 2017; 48:52-108. [PMID: 28816105 DOI: 10.1080/10408444.2017.1351420] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Thiazolidinedione (TZD) drugs used in the treatment of type 2 diabetes mellitus (T2DM) have proven effective in improving insulin sensitivity, hyperglycemia, and lipid metabolism. Though well tolerated by some patients, their mechanism of action as ligands of peroxisome proliferator-activated receptors (PPARs) results in the activation of several pathways in addition to those responsible for glycemic control and lipid homeostasis. These pathways, which include those related to inflammation, bone formation, and cell proliferation, may lead to adverse health outcomes. As treatment with TZDs has been associated with adverse hepatic, cardiovascular, osteological, and carcinogenic events in some studies, the role of TZDs in the treatment of T2DM continues to be debated. At the same time, new therapeutic roles for TZDs are being investigated, with new forms and isoforms currently in the pre-clinical phase for use in the prevention and treatment of some cancers, inflammatory diseases, and other conditions. The aims of this review are to provide an overview of the mechanism(s) of action of TZDs, a review of their safety for use in the treatment of T2DM, and a perspective on their current and future therapeutic roles.
Collapse
Affiliation(s)
- Melissa A Davidson
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada
| | - Donald R Mattison
- b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada
| | - Laurent Azoulay
- d Center for Clinical Epidemiology , Lady Davis Research Institute, Jewish General Hospital , Montreal , Canada.,e Department of Oncology , McGill University , Montreal , Canada
| | - Daniel Krewski
- a Faculty of Health Sciences , University of Ottawa , Ottawa , Canada.,b McLaughlin Centre for Population Health Risk Assessment , Ottawa , Canada.,c Risk Sciences International , Ottawa , Canada.,f Faculty of Medicine , University of Ottawa , Ottawa , Canada
| |
Collapse
|
17
|
Yang Y, Lin S, Wang B, Gu W, Li G. Stem cell therapy for enhancement of bone consolidation in distraction osteogenesis: A contemporary review of experimental studies. Bone Joint Res 2017; 6:385-390. [PMID: 28634158 PMCID: PMC5492338 DOI: 10.1302/2046-3758.66.bjr-2017-0023] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 04/11/2017] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVES Distraction osteogenesis (DO) mobilises bone regenerative potential and avoids the complications of other treatments such as bone graft. The major disadvantage of DO is the length of time required for bone consolidation. Mesenchymal stem cells (MSCs) have been used to promote bone formation with some good results. METHODS We hereby review the published literature on the use of MSCs in promoting bone consolidation during DO. RESULTS Studies differed in animal type (mice, rabbit, dog, sheep), bone type (femur, tibia, skull), DO protocols and cell transplantation methods. CONCLUSION The majority of studies reported that the transplantation of MSCs enhanced bone consolidation or formation in DO. Many questions relating to animal model, DO protocol and cell transplantation regime remain to be further investigated. Clinical trials are needed to test and confirm these findings from animal studies.Cite this article: Y. Yang, S. Lin, B. Wang, W. Gu, G. Li. Stem cell therapy for enhancement of bone consolidation in distraction osteogenesis: A contemporary review of experimental studies. Bone Joint Res 2017;6:385-390. DOI: 10.1302/2046-3758.66.BJR-2017-0023.
Collapse
Affiliation(s)
- Y Yang
- Department of Key Laboratory, Changzhou No.7 People's Hospital, No. 288 Yanling East Road, Changzhou, Jiangsu, China
| | - S Lin
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - B Wang
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| | - W Gu
- Department of Traumatology, Changzhou No.7 People's Hospital, No. 288 Yanling East Road, Changzhou, Jiangsu, China
| | - G Li
- Department of Orthopaedics and Traumatology, Lui Che Woo Institute of Innovative Medicine, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, 30-32 Ngan Shing Street, Prince of Wales Hospital, Shatin, NT, Hong Kong, China
| |
Collapse
|
18
|
Lecka-Czernik B, Stechschulte LA, Czernik PJ, Sherman SB, Huang S, Krings A. Marrow Adipose Tissue: Skeletal Location, Sexual Dimorphism, and Response to Sex Steroid Deficiency. Front Endocrinol (Lausanne) 2017; 8:188. [PMID: 28824548 PMCID: PMC5543291 DOI: 10.3389/fendo.2017.00188] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 07/18/2017] [Indexed: 01/29/2023] Open
Abstract
Marrow adipose tissue (MAT) is unique with respect to origin, metabolism, and function. MAT is characterized with high heterogeneity which correlates with skeletal location and bone metabolism. This fat depot is also highly sensitive to various hormonal, environmental, and pharmacologic cues to which it responds with changes in volume and/or metabolic phenotype. We have demonstrated previously that MAT has characteristics of both white (WAT) and brown (BAT)-like or beige adipose tissue, and that beige phenotype is attenuated with aging and in diabetes. Here, we extended our analysis by comparing MAT phenotype in different locations within a tibia bone of mature C57BL/6 mice and with respect to the presence of sex steroids in males and females. We report that MAT juxtaposed to trabecular bone of proximal tibia (pMAT) is characterized by elevated expression of beige fat markers including Ucp1, HoxC9, Prdm16, Tbx1, and Dio2, when compared with MAT located in distal tibia (dMAT). There is also a difference in tissue organization with adipocytes in proximal tibia being dispersed between trabeculae, while adipocytes in distal tibia being densely packed. Higher trabecular bone mass (BV/TV) in males correlates with lower pMAT volume and higher expression of beige markers in the same location, when compared with females. However, there is no sexual divergence in the volume and transcriptional profile of dMAT. A removal of ovaries in females resulted in decreased cortical bone mass and increased volume of both pMAT and dMAT, as well as volume of gonadal WAT (gWAT). Increase in pMAT volume was associated with marked increase in Fabp4 and Adiponectin expression and relative decrease in beige fat gene markers. A removal of testes in males resulted in cortical and trabecular bone loss and the tendency to increased volume of both pMAT and dMAT, despite a loss of gWAT. Orchiectomy did not affect the expression of white and beige adipocyte gene markers. In conclusion, expression profile of beige adipocyte gene markers correlates with skeletal location of active bone remodeling and higher BV/TV, however bone loss resulted from sex steroid deficiency is not proportional to MAT expansion at the same skeletal location.
Collapse
Affiliation(s)
- Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, Toledo, OH, United States
- Department of Physiology and Pharmacology, University of Toledo Health Sciences Campus, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, Toledo, OH, United States
- *Correspondence: Beata Lecka-Czernik,
| | - Lance A. Stechschulte
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, Toledo, OH, United States
- Center for Diabetes and Endocrine Research, University of Toledo Health Sciences Campus, Toledo, OH, United States
| | - Piotr J. Czernik
- Department of Physiology and Pharmacology, University of Toledo Health Sciences Campus, Toledo, OH, United States
| | - Shermel B. Sherman
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, Toledo, OH, United States
| | - Shilong Huang
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, Toledo, OH, United States
| | - Amrei Krings
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, Toledo, OH, United States
| |
Collapse
|
19
|
Abstract
Distraction osteogenesis biologically resembles fracture healing with distinctive characteristics notably in the distraction phase of osteogenesis. In the latency phase of bone lengthening, like in the inflammatory phase of fracture repair, interleukines are released and act with growth factors released from platelets in the local haematoma, leading to attraction, proliferation and differentiation of mesenchymal stem cells into osteoblasts and other differentiated mesenchymal cells. These in turn produce matrix, collagen fibers and growth factors. A callus containing cells, collagen fibers, osteoid and cartilage matrix is formed. Provided stable fixation, distraction will trigger intramembranous bone formation. As distraction proceeds, the distraction gap develops five distinctive zones with unmineralized bone in the middle, remodelling bone peripherally, and mineralizing bone in between. During consolidation, the high concentration of anabolic growth factors in the regenerate diminishes with time as remodelling takes over to form mature cortical and cancellous bone. Systemic disease, congenital bone deficiencies, medications and substance abuse can influence the quality and quantity of regenerate bone, usually in a negative way. The regenerate bone can be manipulated when needed by using injection of mesenchymal stem cells and platelets, growth factors (BMP-2 and -7), and systemic medications (bisphosphonates and parathyroid hormone). Growth factors and systemic anabolic and antiresorptive drugs are prescribed on special indications, while distraction osteogenesis is not an authorized indication. To some extent, however, these compounds can be used off-label. Use in children presents special problems since growth factors and specific anabolic medications may involve a risk of inducing cancer.
Collapse
Affiliation(s)
- Ivan Hvid
- Section of Pediatric and Reconstructive Orthopaedic Surgery, Department of Orthopaedic Surgery, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway.
| | - Joachim Horn
- Section of Pediatric and Reconstructive Orthopaedic Surgery, Department of Orthopaedic Surgery, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Stefan Huhnstock
- Section of Pediatric and Reconstructive Orthopaedic Surgery, Department of Orthopaedic Surgery, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| | - Harald Steen
- Section of Pediatric and Reconstructive Orthopaedic Surgery, Department of Orthopaedic Surgery, Oslo University Hospital, Rikshospitalet, Sognsvannsveien 20, 0372, Oslo, Norway
| |
Collapse
|
20
|
Stechschulte LA, Ge C, Hinds TD, Sanchez ER, Franceschi RT, Lecka-Czernik B. Protein Phosphatase PP5 Controls Bone Mass and the Negative Effects of Rosiglitazone on Bone through Reciprocal Regulation of PPARγ (Peroxisome Proliferator-activated Receptor γ) and RUNX2 (Runt-related Transcription Factor 2). J Biol Chem 2016; 291:24475-24486. [PMID: 27687725 PMCID: PMC5114402 DOI: 10.1074/jbc.m116.752493] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 09/28/2016] [Indexed: 11/06/2022] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) and runt-related transcription factor 2 (RUNX2) are key regulators of mesenchymal stem cell (MSC) differentiation toward adipocytes and osteoblasts, respectively. Post-translational modifications of these factors determine their activities. Dephosphorylation of PPARγ at Ser-112 is required for its adipocytic activity, whereas phosphorylation of RUNX2 at serine 319 (Ser-319) promotes its osteoblastic activity. Here we show that protein phosphatase 5 (PP5) reciprocally regulates each receptor by targeting each serine. Mice deficient in PP5 phosphatase have increased osteoblast numbers and high bone formation, which results in high bone mass in the appendicular and axial skeleton. This is associated with a substantial decrease in lipid-containing marrow adipocytes. Indeed, in the absence of PP5 the MSC lineage allocation is skewed toward osteoblasts and away from lipid accumulating adipocytes, although an increase in beige adipocyte gene expression is observed. In the presence of rosiglitazone, PP5 translocates to the nucleus, binds to PPARγ and RUNX2, and dephosphorylates both factors, resulting in activation of PPARγ adipocytic and suppression of RUNX2 osteoblastic activities. Moreover, shRNA knockdown of PP5 results in cells refractory to rosiglitazone treatment. Lastly, mice deficient in PP5 are resistant to the negative effects of rosiglitazone on bone, which in wild type animals causes a 50% decrease in trabecular bone mass. In conclusion, PP5 is a unique phosphatase reciprocally regulating PPARγ and RUNX2 activities in marrow MSC.
Collapse
Affiliation(s)
- Lance A Stechschulte
- From the Department of Orthopaedic Surgery,; the Center for Diabetes and Endocrine Research, University of Toledo Health Science Campus, Toledo, Ohio 43614 and
| | - Chunxi Ge
- the Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | | | - Edwin R Sanchez
- the Center for Diabetes and Endocrine Research, University of Toledo Health Science Campus, Toledo, Ohio 43614 and; Physiology and Pharmacology, and
| | - Renny T Franceschi
- the Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan, Ann Arbor, Michigan 48109
| | - Beata Lecka-Czernik
- From the Department of Orthopaedic Surgery,; the Center for Diabetes and Endocrine Research, University of Toledo Health Science Campus, Toledo, Ohio 43614 and; Physiology and Pharmacology, and.
| |
Collapse
|
21
|
La Fontaine J, Chen C, Hunt N, Jude E, Lavery L. Type 2 Diabetes and Metformin Influence on Fracture Healing in an Experimental Rat Model. J Foot Ankle Surg 2016; 55:955-60. [PMID: 27286924 DOI: 10.1053/j.jfas.2016.04.012] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2016] [Indexed: 02/03/2023]
Abstract
Persons with diabetes have a greater incidence of fractures compared with persons without diabetes. However, very little published information is available concerning the deleterious effect of late-stage diabetes on osseous structure and bone healing. The purpose of the present study was to evaluate the role of diabetes on fracture healing in a rat femur repair model. Thirty-six lean and diabetic Zucker rats were subdivided into 3 groups: (1) 12 lean rats as the control group; (2) 12 diabetic rats without blood glucose control (DM group); and (3) 12 diabetic rats treated with 300 mg/kg metformin to reduce the blood glucose levels (DM + Met group). Radiographs were taken every week to determine the incidence of bone repair and delayed union. All the rats were killed at 6 weeks after surgery. In both the sham-operated and the fractured and repaired femurs, significant decreases in the fracture-load/weight and marginal decreases in the fracture-load between the lean and DM groups were found. Metformin treatment significantly reduced the blood glucose and body weight 12 days postoperatively. Furthermore, a decrease in the fracture-load and fracture-load/weight in the repaired femurs was found in the DM + Met group. Diabetes impairs bone fracture healing. Metformin treatment reduces the blood glucose and body weight but had an adverse effect on fracture repair in diabetic rats. Further investigations are needed to reveal the mechanisms responsible for the effects of type 2 diabetes mellitus on bone and bone quality and the effect of medications such as metformin might have in diabetic bone in the presence of neuropathy and vascular disease.
Collapse
Affiliation(s)
- Javier La Fontaine
- Associate Professor, Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX.
| | - Chris Chen
- Assistant Professor, Department of Orthopedics, University of Texas Southwestern Medical Center, Dallas, TX
| | - Nathan Hunt
- Staff Podiatrist, Orthopaedic Center of the Rockies, Fort Collins, CO
| | - Edward Jude
- Consultant Physician/Diabetologist, Tameside General Hospital, Ashton-Under-Lyne, UK
| | - Lawrence Lavery
- Professor, Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
22
|
Yang Y, Luo X, Xie X, Yan F, Chen G, Zhao W, Jiang Z, Fang C, Shen J. Influences of teriparatide administration on marrow fat content in postmenopausal osteopenic women using MR spectroscopy. Climacteric 2016; 19:285-91. [PMID: 26744910 DOI: 10.3109/13697137.2015.1126576] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Objective Teriparatide could induce osteoblast differentiation of mesenchymal stem cells while inhibiting adipocyte differentiation. However, there are significant differences between ex vivo and in vivo models. We aimed to evaluate the impact of teriparatide on marrow and abdominal fat accumulation in postmenopausal osteopenic women. Methods Postmenopausal osteopenic women were randomly assigned to receive teriparatide or placebo for 12 months. Subcutaneous (SAT) and visceral adipose tissue (VAT), marrow fat fraction (MFF), bone density (BMD) and bone biomarkers were measured at baseline, 6 and 12 months. Results At 12 months, mean percentage changes in BMD from baseline were 3.51%, 2.21% and 1.80% at lumbar spine, total hip and femoral neck for the teriparatide group, respectively. Relative to baseline conditions, teriparatide reduced MFF (-3.54% at 6 months; -5.87% at 12 months, all p < 0.01). A significant difference in MFF, but not BMD, was first detected at 6 months (p = 0.012) between groups. MFF was negatively associated with SAT (r = -0.479) and positively associated with VAT (r = 0.531) and VAT/SAT (r = 0.415, all p < 0.05). Teriparatide treatment did not alter abdominal fat composition. Conclusion Teriparatide effectively lowers marrow adiposity but not abdominal fat accumulation in postmenopausal osteopenic women.
Collapse
Affiliation(s)
- Y Yang
- a Department of Radiology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China ;,b Department of Radiology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - X Luo
- a Department of Radiology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China ;,c Department of Radiology , Northern Jiangsu People's Hospital and Clinical Medical College of Yangzhou University , Yangzhou , China
| | - X Xie
- d Department of Radiology , First People's Hospital Affiliated to Shanghai Jiaotong University , Shanghai , China
| | - F Yan
- a Department of Radiology , Ruijin Hospital, Shanghai Jiao Tong University School of Medicine , Shanghai , China
| | - G Chen
- b Department of Radiology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - W Zhao
- b Department of Radiology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - Z Jiang
- b Department of Radiology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - C Fang
- e Department of Endocrinology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| | - J Shen
- b Department of Radiology , The Second Affiliated Hospital of Soochow University , Suzhou , China
| |
Collapse
|
23
|
Pagnotti GM, Styner M. Exercise Regulation of Marrow Adipose Tissue. Front Endocrinol (Lausanne) 2016; 7:94. [PMID: 27471493 PMCID: PMC4943947 DOI: 10.3389/fendo.2016.00094] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Accepted: 07/04/2016] [Indexed: 12/20/2022] Open
Abstract
Despite association with low bone density and skeletal fractures, marrow adipose tissue (MAT) remains poorly understood. The marrow adipocyte originates from the mesenchymal stem cell (MSC) pool that also gives rise to osteoblasts, chondrocytes, and myocytes, among other cell types. To date, the presence of MAT has been attributed to preferential biasing of MSC into the adipocyte rather than osteoblast lineage, thus negatively impacting bone formation. Here, we focus on understanding the physiology of MAT in the setting of exercise, dietary interventions, and pharmacologic agents that alter fat metabolism. The beneficial effect of exercise on musculoskeletal strength is known: exercise induces bone formation, encourages growth of skeletally supportive tissues, inhibits bone resorption, and alters skeletal architecture through direct and indirect effects on a multiplicity of cells involved in skeletal adaptation. MAT is less well studied due to the lack of reproducible quantification techniques. In recent work, osmium-based 3D quantification shows a robust response of MAT to both dietary and exercise intervention in that MAT is elevated in response to high-fat diet and can be suppressed following daily exercise. Exercise-induced bone formation correlates with suppression of MAT, such that exercise effects might be due to either calorie expenditure from this depot or from mechanical biasing of MSC lineage away from fat and toward bone, or a combination thereof. Following treatment with the anti-diabetes drug rosiglitazone - a PPARγ-agonist known to increase MAT and fracture risk - mice demonstrate a fivefold higher femur MAT volume compared to the controls. In addition to preventing MAT accumulation in control mice, exercise intervention significantly lowers MAT accumulation in rosiglitazone-treated mice. Importantly, exercise induction of trabecular bone volume is unhindered by rosiglitazone. Thus, despite rosiglitazone augmentation of MAT, exercise significantly suppresses MAT volume and induces bone formation. That exercise can both suppress MAT volume and increase bone quantity, notwithstanding the skeletal harm induced by rosiglitazone, underscores exercise as a powerful regulator of bone remodeling, encouraging marrow stem cells toward the osteogenic lineage to fulfill an adaptive need for bone formation. Thus, exercise represents an effective strategy to mitigate the deleterious effects of overeating and iatrogenic etiologies on bone and fat.
Collapse
Affiliation(s)
- Gabriel M. Pagnotti
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Maya Styner
- Department of Medicine, University of North Carolina, Chapel Hill, NC, USA
- *Correspondence: Maya Styner,
| |
Collapse
|
24
|
Picke AK, Gordaliza Alaguero I, Campbell GM, Glüer CC, Salbach-Hirsch J, Rauner M, Hofbauer LC, Hofbauer C. Bone defect regeneration and cortical bone parameters of type 2 diabetic rats are improved by insulin therapy. Bone 2016; 82:108-15. [PMID: 26055107 DOI: 10.1016/j.bone.2015.06.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/27/2015] [Accepted: 06/02/2015] [Indexed: 12/26/2022]
Abstract
Zucker Diabetic Fatty (ZDF) rats represent an established model of type 2 diabetes mellitus (T2DM) and display several features of human diabetic bone disease, including impaired osteoblast function, decreased bone strength, and delayed bone healing. Here, we determined whether glycemic control by insulin treatment prevents skeletal complications associated with diabetes. Subcritical femur defects were created in diabetic (fa/fa) and non-diabetic (+/+) ZDF rats. Diabetic rats were treated once daily with long-lasting insulin glargin for 12weeks for glycemic control. Insulin treatment successfully maintained serum levels of glycated hemoglobin, while untreated diabetic rats showed a 2-fold increase. Trabecular and cortical bone mass measured by μCT were decreased in diabetic rats. Insulin treatment increased bone mass of the cortical, but not of the trabecular bone compartment. Dynamic histomorphometry revealed a lower bone formation rate at the trabecular and periosteal cortical bone in diabetic animals and decreased serum procollagen type 1 N-terminal propeptide (P1NP, -49%) levels. Insulin treatment partially improved these parameters. In T2DM, serum levels of tartrate-resistant acid phosphatase (TRAP, +32%) and C-terminal telopeptide (CTX, +49%) were increased. Insulin treatment further elevated TRAP levels, but did not affect CTX levels. While diabetes impaired bone defect healing, glycemic control with insulin fully reversed these negative effects. In conclusion, insulin treatment reversed the adverse effects of T2DM on bone defect regeneration in rats mainly by improving osteoblast function and bone formation. This article is part of a Special Issue entitled Bone and diabetes.
Collapse
Affiliation(s)
- A-K Picke
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Germany
| | - I Gordaliza Alaguero
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Germany
| | - G M Campbell
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, Christian-Albrechts-Universität zu Kiel, Germany
| | - C-C Glüer
- Section Biomedical Imaging, MOIN CC, Department of Radiology and Neuroradiology, Christian-Albrechts-Universität zu Kiel, Germany
| | - J Salbach-Hirsch
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Germany
| | - M Rauner
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Germany
| | - L C Hofbauer
- Division of Endocrinology, Diabetes, and Metabolic Bone Diseases, Department of Medicine III, Technische Universität Dresden, Germany; DFG Research Center and Cluster of Excellence for Regenerative Therapies, Technische Universität Dresden, Germany
| | - C Hofbauer
- Department of Orthopedics, Technische Universität Dresden, Germany.
| |
Collapse
|
25
|
Abstract
Diabetes is associated with a number of lower extremity orthopedic conditions and complications including fractures, Charcot neuroarthropathy, plantar ulcers, and infection. These complications are of significant clinical concern in terms of morbidity, mortality, and socioeconomic costs. A review of each condition is discussed, with particular emphasis on the clinical importance, diagnostic considerations, and orthopedic treatment recommendations. The goal of the article is to provide a clinical picture of the challenges that orthopedic surgeons confront, and highlight the need for specific clinical guidelines in diabetic patients.
Collapse
Affiliation(s)
- Daniel J Gehling
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH 43614, United States.
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH 43614, United States; Department of Physiology and Pharmacology, University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH 43614, United States
| | - Nabil A Ebraheim
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, 3000 Arlington Avenue, Toledo, OH 43614, United States
| |
Collapse
|
26
|
Styner M, Pagnotti GM, Galior K, Wu X, Thompson WR, Uzer G, Sen B, Xie Z, Horowitz MC, Styner MA, Rubin C, Rubin J. Exercise Regulation of Marrow Fat in the Setting of PPARγ Agonist Treatment in Female C57BL/6 Mice. Endocrinology 2015; 156:2753-61. [PMID: 26052898 PMCID: PMC4511140 DOI: 10.1210/en.2015-1213] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The contribution of marrow adipose tissue (MAT) to skeletal fragility is poorly understood. Peroxisome proliferator-activated receptor (PPAR)γ agonists, associated with increased fractures in diabetic patients, increase MAT. Here, we asked whether exercise could limit the MAT accrual and increase bone formation in the setting of PPARγ agonist treatment. Eight-week-old female C57BL/6 mice were treated with 20-mg/kg · d rosiglitazone (Rosi) and compared with control (CTL) animals. Exercise groups ran 12 km/d when provided access to running wheels (CTL exercise [CTL-E], Rosi-E). After 6 weeks, femoral MAT (volume of lipid binder osmium) and tibial bone morphology were assessed by microcomputer tomography. Rosi was associated with 40% higher femur MAT volume compared with CTL (P < .0001). Exercise suppressed MAT volume by half in CTL-E mice compared with CTL (P < .01) and 19% in Rosi-E compared with Rosi (P < .0001). Rosi treatment increased fat markers perilipin and fatty acid synthase mRNA by 4-fold (P < .01). Exercise was associated with increased uncoupling protein 1 mRNA expression in both CTL-E and Rosi-E groups (P < .05), suggestive of increased brown fat. Rosi increased cortical porosity (P < .0001) but did not significantly impact trabecular or cortical bone quantity. Importantly, exercise induction of trabecular bone volume was not prevented by Rosi (CTL-E 21% > CTL, P < .05; Rosi-E 26% > Rosi, P < .01). In summary, despite the Rosi induction of MAT extending well into the femoral diaphysis, exercise was able to significantly suppress MAT volume and induce bone formation. Our results suggest that the impact of PPARγ agonists on bone and marrow health can be partially mitigated by exercise.
Collapse
Affiliation(s)
- Maya Styner
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Gabriel M Pagnotti
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Kornelia Galior
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Xin Wu
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - William R Thompson
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Gunes Uzer
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Buer Sen
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Zhihui Xie
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Mark C Horowitz
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Martin A Styner
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Clinton Rubin
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| | - Janet Rubin
- Department of Medicine (M.S., K.G., X.W., G.U., B.S., Z.X., J.R.), University of North Carolina, Chapel Hill, North Carolina; Department of Physical Therapy (W.R.T.), Indiana University, Indianapolis, Indiana; Department of Computer Science (M.A.S.), University of North Carolina, Chapel Hill, North Carolina; Department of Psychiatry (M.A.S.), Image Analysis, University of North Carolina, Chapel Hill, North Carolina; Department of Biomedical Engineering (G.M.P., C.R.), Stony Brook University, Stony Brook, New York; and Department of Orthopedics and Rehabilitation (M.C.H.), Yale University, New Haven, Connecticut
| |
Collapse
|
27
|
Lecka-Czernik B, Stechschulte LA, Czernik PJ, Dowling AR. High bone mass in adult mice with diet-induced obesity results from a combination of initial increase in bone mass followed by attenuation in bone formation; implications for high bone mass and decreased bone quality in obesity. Mol Cell Endocrinol 2015; 410:35-41. [PMID: 25576855 DOI: 10.1016/j.mce.2015.01.001] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Revised: 12/23/2014] [Accepted: 01/02/2015] [Indexed: 11/22/2022]
Abstract
Obesity is generally recognized as a condition which positively influences bone mass and bone mineral density (BMD). Positive effect of high body mass index (BMI) on bone has been recognized as a result of increased mechanical loading exerted on the skeleton. However, epidemiologic studies indicate that obesity is associated with increased incidence of fractures. The results presented here offer a new perspective regarding the mechanisms which may be responsible for the increase of bone mass and concurrent decrease in bone quality. Two groups of 12 week old C57BL/6 males were fed either high fat diet (HFD) or regular diet (RD) for 11 weeks. Metabolic profile, bone parameters and gene expression were assessed in these groups at the end of the experiment. Additionally, bone status was evaluated in a third group of 12 week old animals corresponding to animals at the start of the feeding period. Administration of HFD resulted in development of a diet-induced obesity (DIO), glucose intolerance, alteration in energy metabolism, and impairment in WAT function, as compared to the age-matched control animals fed RD. The expression of adiponectin, FABP4/aP2, DIO2 and FoxC2 were decreased in WAT of DIO animals, as well as transcript levels for IGFBP2, the cytokine regulating both energy metabolism and bone mass. At the end of experiment, DIO mice had higher bone mass than both control groups on RD, however they had decreased bone formation, as assessed by calcein labeling, and increased marrow adipocyte content. This study suggests that the bone mass acquired in obesity is a result of a two-phase process. First phase would consist of either beneficial effect of fat expansion to increase bone mass by increased mechanical loading and/or increased production of bone anabolic adipokines and/or nutritional effect of fatty acids. This is followed by a second phase characterized by decreased bone formation and bone turnover resulting from development of metabolic impairment.
Collapse
Affiliation(s)
- B Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA; Department of Physiology and Pharmacology, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research (CeDER), University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA.
| | - L A Stechschulte
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA; Center for Diabetes and Endocrine Research (CeDER), University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - P J Czernik
- Department of Orthopaedic Surgery, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA
| | - A R Dowling
- Department of Physiology and Pharmacology, University of Toledo Health Sciences Campus, 3000 Arlington Avenue, Toledo, OH 43614, USA
| |
Collapse
|
28
|
Chen X, Hausman BS, Luo G, Zhou G, Murakami S, Rubin J, Greenfield EM. Protein kinase inhibitor γ reciprocally regulates osteoblast and adipocyte differentiation by downregulating leukemia inhibitory factor. Stem Cells 2015; 31:2789-99. [PMID: 23963683 DOI: 10.1002/stem.1524] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Revised: 07/22/2013] [Accepted: 07/22/2013] [Indexed: 12/26/2022]
Abstract
The protein kinase inhibitor (Pki) gene family inactivates nuclear protein kinase A (PKA) and terminates PKA-induced gene expression. We previously showed that Pkig is the primary family member expressed in osteoblasts and that Pkig knockdown increases the effects of parathyroid hormone and isoproterenol on PKA activation, gene expression, and inhibition of apoptosis. Here, we determined whether endogenous levels of Pkig regulate osteoblast differentiation. Pkig is the primary family member in murine embryonic fibroblasts (MEFs), murine marrow-derived mesenchymal stem cells, and human mesenchymal stem cells. Pkig deletion increased forskolin-dependent nuclear PKA activation and gene expression and Pkig deletion or knockdown increased osteoblast differentiation. PKA signaling is known to stimulate adipogenesis; however, adipogenesis and osteogenesis are often reciprocally regulated. We found that the reciprocal regulation predominates over the direct effects of PKA since adipogenesis was decreased by Pkig deletion or knockdown. Pkig deletion or knockdown also simultaneously increased osteogenesis and decreased adipogenesis in mixed osteogenic/adipogenic medium. Pkig deletion increased PKA-induced expression of leukemia inhibitory factor (Lif) mRNA and LIF protein. LIF neutralizing antibodies inhibited the effects on osteogenesis and adipogenesis of either Pkig deletion in MEFs or PKIγ knockdown in both murine and human mesenchymal stem cells. Collectively, our results show that endogenous levels of Pkig reciprocally regulate osteoblast and adipocyte differentiation and that this reciprocal regulation is mediated in part by LIF. Stem Cells 2013;31:2789-2799.
Collapse
Affiliation(s)
- Xin Chen
- Department of Orthopaedics, University of North Carolina, Chapel Hill, North Carolina, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Ackerson RM, Shum LC, Berry AR, Bucknell AL, King KB. In vivo model to measure bone repair efficacy of nanoparticle-based drug delivery. Orthopedics 2014; 37:e707-11. [PMID: 25102506 DOI: 10.3928/01477447-20140728-56] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/30/2014] [Indexed: 02/03/2023]
Abstract
Bone repair required for successful arthroplasty can be compromised in patients with comorbid conditions, such as osteoporosis, diabetes mellitus, and chronic kidney disease. Biological compounds have been proposed to promote bone health and repair. The authors have designed a new animal model for testing bone promoting compounds in the in vivo environment. For initial validation of this model, they used a synthetic agonist of a nuclear receptor, liver X receptor, which has been postulated to play a regulatory role in modulating bone growth. A distal femoral unicortical osteotomy was surgically created on skeletally mature C57Bl/6 male and female mice. A nanoparticle carrier delivery system was used to directly introduce N,N-dimethyl-3β-hydroxycholenamide into the osteotomy. At 35 days post-procedure, the femora were harvested and specimens were obtained for histologic processing and qualitative analysis. The results indicate that the carrier nanoparticles entered the osteotomy defect. Results also indicate that bone repair occurred, although significant differences between groups were not detected in the current study. This study validates the mouse model for testing bone repair promoting compounds. This model can be combined with transgenic or other mouse models to simulate problematic bone repair environments, can be used with a variety of drug carriers, and can test many types of interventional compounds to evaluate potential orthopedic therapeutic applications.
Collapse
|
30
|
Lecka-Czernik B, Stechschulte LA. Bone and fat: a relationship of different shades. Arch Biochem Biophys 2014; 561:124-9. [PMID: 24956594 DOI: 10.1016/j.abb.2014.06.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 06/10/2014] [Accepted: 06/12/2014] [Indexed: 12/21/2022]
Abstract
Environmental and behavioral changes which occurred over the last century led simultaneously to a remarkable increase in human lifespan and to the development of health problems associated with functional impairment of organs either regulating or dependent on balanced energy metabolism. Diseases such as diabetes, obesity and osteoporosis are prevalent in our society and pose major challenges with respect to the overall health and economy. Therefore, better understanding of regulatory axes between bone and fat may provide the basis for development of strategies which will treat these diseases simultaneously and improve health and life quality of elderly.
Collapse
Affiliation(s)
- Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo Health Science Campus, Toledo, OH 43614, United States; Department of Physiology and Pharmacology, University of Toledo Health Science Campus, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, University of Toledo Health Science Campus, Toledo, OH 43614, United States.
| | - Lance A Stechschulte
- Department of Orthopaedic Surgery, University of Toledo Health Science Campus, Toledo, OH 43614, United States; Center for Diabetes and Endocrine Research, University of Toledo Health Science Campus, Toledo, OH 43614, United States
| |
Collapse
|
31
|
Kolli V, Stechschulte LA, Dowling AR, Rahman S, Czernik PJ, Lecka-Czernik B. Partial agonist, telmisartan, maintains PPARγ serine 112 phosphorylation, and does not affect osteoblast differentiation and bone mass. PLoS One 2014; 9:e96323. [PMID: 24810249 PMCID: PMC4014504 DOI: 10.1371/journal.pone.0096323] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 04/07/2014] [Indexed: 11/30/2022] Open
Abstract
Peroxisome proliferator activated receptor gamma (PPARγ) controls both glucose metabolism and an allocation of marrow mesenchymal stem cells (MSCs) toward osteoblast and adipocyte lineages. Its activity is determined by interaction with a ligand which directs posttranscriptional modifications of PPARγ protein including dephosphorylation of Ser112 and Ser273, which results in acquiring of pro-adipocytic and insulin-sensitizing activities, respectively. PPARγ full agonist TZD rosiglitazone (ROSI) decreases phosphorylation of both Ser112 and Ser273 and its prolonged use causes bone loss in part due to diversion of MSCs differentiation from osteoblastic toward adipocytic lineage. Telmisartan (TEL), an anti-hypertensive drug from the class of angiotensin receptor blockers, also acts as a partial PPARγ agonist with insulin-sensitizing and a weak pro-adipocytic activity. TEL decreased S273pPPARγ and did not affect S112pPPARγ levels in a model of marrow MSC differentiation, U-33/γ2 cells. In contrast to ROSI, TEL did not affect osteoblast phenotype and actively blocked ROSI-induced anti-osteoblastic activity and dephosphorylation of S112pPPARγ. The effect of TEL on bone was tested side-by-side with ROSI. In contrast to ROSI, TEL administration did not affect bone mass and bone biomechanical properties measured by micro-indentation method and did not induce fat accumulation in bone, and it partially protected from ROSI-induced bone loss. In addition, TEL induced “browning” of epididymal white adipose tissue marked by increased expression of UCP1, FoxC2, Wnt10b and IGFBP2 and increased overall energy expenditure. These studies point to the complexity of mechanisms by which PPARγ acquires anti-osteoblastic and pro-adipocytic activities and suggest an importance of Ser112 phosphorylation status as being a part of the mechanism regulating this process. These studies showed that TEL acts as a full PPARγ agonist for insulin-sensitizing activity and as a partial agonist/partial antagonist for pro-adipocytic and anti-osteoblastic activities. They also suggest a relationship between PPARγ fat “browning” activity and a lack of anti-osteoblastic activity.
Collapse
Affiliation(s)
- Vipula Kolli
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Lance A. Stechschulte
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Abigail R. Dowling
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Sima Rahman
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Piotr J. Czernik
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, Toledo, Ohio, United States of America
| | - Beata Lecka-Czernik
- Department of Orthopaedic Surgery, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- Center for Diabetes and Endocrine Research, University of Toledo College of Medicine, Toledo, Ohio, United States of America
- * E-mail:
| |
Collapse
|
32
|
Ruan J, Trotter TN, Nan L, Luo R, Javed A, Sanderson RD, Suva LJ, Yang Y. Heparanase inhibits osteoblastogenesis and shifts bone marrow progenitor cell fate in myeloma bone disease. Bone 2013; 57:10-7. [PMID: 23895995 PMCID: PMC3786009 DOI: 10.1016/j.bone.2013.07.024] [Citation(s) in RCA: 39] [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: 04/15/2013] [Revised: 06/15/2013] [Accepted: 07/19/2013] [Indexed: 11/25/2022]
Abstract
A major cause of morbidity in patients with multiple myeloma is the development and progression of bone disease. Myeloma bone disease is characterized by rampant osteolysis in the presence of absent or diminished bone formation. Heparanase, an enzyme that acts both at the cell-surface and within the extracellular matrix to degrade polymeric heparan sulfate chains, is upregulated in a variety of human cancers including multiple myeloma. We and others have shown that heparanase enhances osteoclastogenesis and bone loss. However, increased osteolysis is only one element of the spectrum of myeloma bone disease. In the present study, we hypothesized that heparanase would also affect mesenchymal cells in the bone microenvironment and investigated the effect of heparanase on the differentiation of osteoblast/stromal lineage cells. Using a combination of molecular, biochemical, cellular and in vivo approaches, we demonstrated that heparanase significantly inhibited osteoblast differentiation and mineralization, and reduced bone formation in vivo. In addition, heparanase shifts the differentiation potential of osteoblast progenitors from osteoblastogenesis to adipogenesis. Mechanistically, this shift in cell fate is due, at least in part, to heparanase-enhanced production and secretion of the Wnt signaling pathway inhibitor DKK1 by both osteoblast progenitors and myeloma cells. Collectively, these data provide important new insights into the role of heparanase in all aspects of myeloma bone disease and strongly support the use of heparanase inhibitors in the treatment of multiple myeloma.
Collapse
Affiliation(s)
- Jian Ruan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
- Department of Oncology, Nanfang Hospital, Guangzhou, China
| | - Timothy N. Trotter
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Li Nan
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
| | - Rongcheng Luo
- Department of Oncology, Nanfang Hospital, Guangzhou, China
| | - Amjad Javed
- Comprehensive Cancer Center and the Center for Metabolic Bone Disease, University of Alabama at Birmingham, Birmingham, AL
- Department of Oral and Maxillofacial Surgery, School of Dentistry, University of Alabama at Birmingham, Birmingham, AL
| | - Ralph D. Sanderson
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Cancer Center and the Center for Metabolic Bone Disease, University of Alabama at Birmingham, Birmingham, AL
| | - Larry J. Suva
- Department of Orthopaedic Surgery, Center for Orthopaedic Research, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Yang Yang
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL
- Comprehensive Cancer Center and the Center for Metabolic Bone Disease, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
33
|
Rahman S, Lu Y, Czernik PJ, Rosen CJ, Enerback S, Lecka-Czernik B. Inducible brown adipose tissue, or beige fat, is anabolic for the skeleton. Endocrinology 2013; 154:2687-701. [PMID: 23696565 PMCID: PMC3713216 DOI: 10.1210/en.2012-2162] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
It is known that insulin resistance and type 2 diabetes mellitus are associated with increased fractures and that brown adipose tissue (BAT) counteracts many if not all of the symptoms associated with type 2 diabetes. By the use of FoxC2(AD)(+/Tg) mice, a well-established model for induction of BAT, or beige fat, we present data extending the beneficial action of beige fat to also include a positive effect on bone. FoxC2(AD)(+/Tg) mice are lean and insulin-sensitive and have high bone mass due to increased bone formation associated with high bone turnover. Inducible BAT is linked to activation of endosteal osteoblasts whereas osteocytes have decreased expression of the Sost transcript encoding sclerostin and elevated expression of Rankl. Conditioned media (CM) collected from forkhead box c2 (FOXC2)-induced beige adipocytes activated the osteoblast phenotype and increased levels of phospho-AKT and β-catenin in recipient cells. In osteocytes, the same media decreased Sost expression. Immunodepletion of CM with antibodies against wingless related MMTV integration site 10b (WNT10b) and insulin-like growth factor binding protein 2 (IGFBP2) resulted in the loss of pro-osteoblastic activity, and the loss of increase in the levels of phospho-AKT and β-catenin. Conversely, CM derived from cells overexpressing IGFBP2 or WNT10b restored osteoblastic activity in recipient cells. In conclusion, beige fat secretes endocrine/paracrine activity that is beneficial for the skeleton.
Collapse
Affiliation(s)
- Sima Rahman
- Departments of Orthopaedic Surgery, University ofToledo Health Sciences Campus, Toledo, OH 43614, USA
| | | | | | | | | | | |
Collapse
|
34
|
Yau H, Rivera K, Lomonaco R, Cusi K. The future of thiazolidinedione therapy in the management of type 2 diabetes mellitus. Curr Diab Rep 2013; 13:329-41. [PMID: 23625197 DOI: 10.1007/s11892-013-0378-8] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Since their approval, thiazolidinediones (TZDs) have been used extensively as insulin-sensitizers for the management of type 2 diabetes mellitus (T2DM). Activation of peroxisomal proliferator-activated receptor gamma (PPARγ) nuclear receptors by TZDs leads to a vast spectrum of metabolic and antiinflammatory effects. In the past decade, clinicians and scientists across the fields of metabolism, diabetes, liver disease (NAFLD), atherosclerosis, inflammation, infertility, and even cancer have had high hopes about the potential for TZDs to treat many of these diseases. However, an increasing awareness about undesirable "off-target" effects of TZDs have made us rethink their role and be more cautious about the long-term benefits and risks related to their use. This review examines the most relevant work on the benefits and risks associated with TZD treatment, with a focus on the only PPARγ agonist currently available (pioglitazone), aiming to offer the reader a balanced overview about the current and future role of TZDs in the management of insulin-resistant states and T2DM.
Collapse
Affiliation(s)
- Hanford Yau
- Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, The University of Florida North Florida/South Georgia Veterans Health System, Gainesville, Florida, USA
| | | | | | | |
Collapse
|