151
|
Wu PH, Glerup RI, Svensson MHS, Eriksson N, Christensen JH, Linde T, Ljunggren Ö, Fellström B. Osteoprotegerin predicts cardiovascular events in patients treated with hemodialysis. Nephrol Dial Transplant 2021; 37:1162-1170. [PMID: 34086939 DOI: 10.1093/ndt/gfab192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Disturbances in bone mineral metabolism are associated with increased mortality and cardiovascular events (CVEs). However, the association between bone-associated protein biomarkers, mortality, and CVEs independent of cytokine activation remains unknown. This study aimed to investigate bone-associated protein biomarkers, and the association with inflammatory cytokines, and cardiovascular outcomes. METHODS This prospective study enrolled hemodialysis (HD) patients in Denmark between December 2010 and March 2011. Using a proximity extension proteomics assay, nine bone-associated proteins were examined: cathepsin D (CTSD), cathepsin L1 (CTSL1), dickkopf-related protein 1 (Dkk-1), fibroblast growth factor 23 (FGF-23), leptin, osteoprotegerin (OPG), receptor activator of nuclear factor kappa-Β ligand (RANKL), TNF-related apoptosis-inducing ligand (TRAIL), and TNF-related apoptosis-inducing ligand receptor 2 (TRAIL-R2). The importance of the bone-associated protein markers was evaluated by a random forest algorithm (RF). The association between bone-associated proteins with all-cause death, cardiovascular death, and CVEs was analyzed in multivariable Cox models adjusted for age, gender, comorbidities, laboratory data, and dialysis duration. RESULTS We enrolled 331 patients (63.7% men; mean [SD] age, 65 [14.6] years) in a prospective cohort study with five years follow-up. When adjusting for confounders, CTSL1 remained associated with all-cause death, and four biomarkers were associated with CVE. However, the association between bone markers and the outcomes was attenuated after adjusting for inflammatory proteins, and just OPG remained associated with CVE in the adjusted model. Evaluating the importance of bone markers by RF, OPG was the most important marker related to CVEs. OPG also improved the prediction of CVE when added clinical information alone in integrated discrimination improvement and net reclassification improvement analyses. DISCUSSION OPG, a well-known bone biomarker, was associated with CVEs independent of cytokine activity. In contrast, the association between CVEs and the remaining three bone-associated proteins (TRAIL-R2, CTSD, and CTSL1) was affected by cytokine inflammation activity.
Collapse
Affiliation(s)
- Ping-Hsun Wu
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden.,Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.,Division of Nephrology, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Rie Io Glerup
- Department of Nephrology, Aalborg University Hospital, Aalborg, Denmark
| | - My Hanna Sofia Svensson
- Division of Medicine, Department of Nephrology, Akershus University Hospital, Oslo, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Niclas Eriksson
- Uppsala University Uppsala Clinical Research Center, and Department of Medical Sciences Uppsala, Sweden
| | | | - Torbjörn Linde
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Östen Ljunggren
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Bengt Fellström
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
152
|
Hamoudi D, Bouredji Z, Marcadet L, Yagita H, Landry LB, Argaw A, Frenette J. Muscle weakness and selective muscle atrophy in osteoprotegerin-deficient mice. Hum Mol Genet 2021; 29:483-494. [PMID: 31943048 DOI: 10.1093/hmg/ddz312] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/07/2019] [Accepted: 12/07/2019] [Indexed: 01/07/2023] Open
Abstract
Bone and muscle are tightly coupled and form a functional unit under normal conditions. The receptor-activator of nuclear factor κB/receptor-activator of nuclear factor κB ligand/osteoprotegerin (RANK/RANKL/OPG) triad plays a crucial role in bone remodeling. RANKL inhibition by OPG prevents osteoporosis. In contrast, the absence of OPG results in elevated serum RANKL and early onset osteoporosis. However, the impacts of OPG deletion on muscle structure and function are unknown. Our results showed that 1-, 3- and 5-month-old Opg-/- mice have reduced tibial and femoral bone biomechanical properties and higher levels of circulating RANKL. OPG-deficient mice displayed reduced locomotor activity and signs of muscle weakness at 5 months of age. Furthermore, OPG deficiency did not affect the skeletal muscles in 1- and 3-month-old mice. However, it impaired fast-twitch EDL but not slow-twitch Sol muscles in 5-month-old Opg-/- mice. Moreover, 5-month-old Opg-/- mice exhibited selective atrophy of fast-twitch-type IIb myofibers, with increased expression of atrophic proteins such as NF-kB, atrogin-1 and MuRF-1. We used an in vitro model to show that RANKL-stimulated C2C12 myotubes significantly increased the expression of NF-kB, atrogin-1 and MuRF-1. A 2-month anti-RANKL treatment starting at 3 months of age in Opg-/- mice improved voluntary activity, the ex vivo maximum specific force (sP0) of EDL muscles, and whole limb grip force performance and rescued the biomechanical properties of bone. In conclusion, the deletion of OPG and the disruption of the RANKL/OPG balance induced osteoporosis as well as the selective weakness and atrophy of the powerful fast-twitch IIb myofibers, which was partly alleviated by an anti-RANKL treatment.
Collapse
Affiliation(s)
- Dounia Hamoudi
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - Zineb Bouredji
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - Laetitia Marcadet
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - Hideo Yagita
- Department of Immunology, School of Medicine, Juntendo University, Tokyo, Japan
| | - Louis-Bénédict Landry
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - Anteneh Argaw
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada
| | - Jérôme Frenette
- Centre Hospitalier Universitaire de Québec, Centre de Recherche du Centre Hospitalier de l'Université Laval (CHUQ-CHUL), Axe Neurosciences, Université Laval, Quebec City, QC G1V 4G2, Canada.,Département de Réadaptation, Faculté de Médecine, Université Laval, Quebec City, QC G1V 0A6, Canada
| |
Collapse
|
153
|
Tsukasaki M, Asano T, Muro R, Huynh NCN, Komatsu N, Okamoto K, Nakano K, Okamura T, Nitta T, Takayanagi H. OPG Production Matters Where It Happened. Cell Rep 2021; 32:108124. [PMID: 32905763 DOI: 10.1016/j.celrep.2020.108124] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 08/05/2020] [Accepted: 08/18/2020] [Indexed: 12/20/2022] Open
Abstract
Osteoprotegerin (OPG) is a circulating decoy receptor for RANKL, a multifunctional cytokine essential for the differentiation of tissue-specific cells in bone and immune systems such as osteoclasts, medullary thymic epithelial cells (mTECs), and intestinal microfold cells (M cells). However, it is unknown whether OPG functions only at the production site or circulates to other tissues acting in an endocrine fashion. Here we explore the cellular source of OPG by generating OPG-floxed mice and show that locally produced OPG, rather than circulating OPG, is crucial for bone and immune homeostasis. Deletion of OPG in osteoblastic cells leads to severe osteopenia without affecting serum OPG. Deletion of locally produced OPG increases mTEC and M cell numbers while retaining the normal serum OPG level. This study shows that OPG limits its functions within the tissue where it was produced, illuminating the importance of local regulation of the RANKL system.
Collapse
Affiliation(s)
- Masayuki Tsukasaki
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Tatsuo Asano
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Ryunosuke Muro
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Nam Cong-Nhat Huynh
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Noriko Komatsu
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Kazuo Okamoto
- Department of Osteoimmunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Kenta Nakano
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, 162-8655 Tokyo, Japan
| | - Tadashi Okamura
- Department of Laboratory Animal Medicine, Research Institute, National Center for Global Health and Medicine, 1-21-1, Toyama, Shinjuku-ku, 162-8655 Tokyo, Japan
| | - Takeshi Nitta
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan
| | - Hiroshi Takayanagi
- Department of Immunology, Graduate School of Medicine and Faculty of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, 113-0033 Tokyo, Japan.
| |
Collapse
|
154
|
Local Production of Osteoprotegerin by Osteoblasts Suppresses Bone Resorption. Cell Rep 2021; 32:108052. [PMID: 32905775 PMCID: PMC7493998 DOI: 10.1016/j.celrep.2020.108052] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 06/15/2020] [Accepted: 07/29/2020] [Indexed: 11/21/2022] Open
Abstract
Osteoprotegerin (OPG) inhibits the ability of receptor activator of nuclear factor κB (NF-κB) ligand (RANKL) to stimulate the differentiation, activity, and survival of bone-resorbing osteoclasts. Genetic studies in mice show that osteocytes are an important source of RANKL, but the cellular sources of OPG are unclear. We use conditional deletion of Tnfrsf11b, which encodes OPG, from different cell populations to identify functionally relevant sources of OPG in mice. Deletion from B lymphocytes and osteocytes, two cell types commonly thought to supply OPG, has little or no impact on bone mass. By contrast, deletion of Tnfrsf11b from osteoblasts increases bone resorption and reduces bone mass to an extent similar to germline deletion, demonstrating that osteoblasts are an essential source of OPG. These results suggest that, in addition to producing new bone matrix, osteoblasts also play an active role in terminating the resorption phase of the bone remodeling cycle by suppressing RANKL activity.
Collapse
|
155
|
Herrmann J, Gummi MR, Xia M, van der Giet M, Tölle M, Schuchardt M. Vascular Calcification in Rodent Models-Keeping Track with an Extented Method Assortment. BIOLOGY 2021; 10:biology10060459. [PMID: 34067504 PMCID: PMC8224561 DOI: 10.3390/biology10060459] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/12/2021] [Accepted: 05/20/2021] [Indexed: 02/07/2023]
Abstract
Simple Summary Arterial vessel diseases are the leading cause of death in the elderly and their accelerated pathogenesis is responsible for premature death in patients with chronic renal failure. Since no functioning therapy concepts exist so far, the identification of the main signaling pathways is of current research interest. To develop therapeutic concepts, different experimental rodent models are needed, which should be subject to the 3R principle of Russel and Burch: “Replace, Reduce and Refine”. This review aims to summarize the current available experimental rodent models for studying vascular calcification and their quantification methods. Abstract Vascular calcification is a multifaceted disease and a significant contributor to cardiovascular morbidity and mortality. The calcification deposits in the vessel wall can vary in size and localization. Various pathophysiological pathways may be involved in disease progression. With respect to the calcification diversity, a great number of research models and detection methods have been established in basic research, relying mostly on rodent models. The aim of this review is to provide an overview of the currently available rodent models and quantification methods for vascular calcification, emphasizing animal burden and assessing prospects to use available methods in a way to address the 3R principles of Russel and Burch: “Replace, Reduce and Refine”.
Collapse
Affiliation(s)
- Jaqueline Herrmann
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
- Department of Chemistry, Biochemistry and Pharmacy, Freie Universität Berlin, Königin-Luise-Straße 2+4, 14195 Berlin, Germany
| | - Manasa Reddy Gummi
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Mengdi Xia
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Markus van der Giet
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Markus Tölle
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
| | - Mirjam Schuchardt
- Department of Nephrology and Medical Intensive Care, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Hindenburgdamm 30, 12203 Berlin, Germany; (J.H.); (M.R.G.); (M.X.); (M.v.d.G.); (M.T.)
- Correspondence: ; Tel.: +49-30-450-514-690
| |
Collapse
|
156
|
Kutikhin AG, Feenstra L, Kostyunin AE, Yuzhalin AE, Hillebrands JL, Krenning G. Calciprotein Particles: Balancing Mineral Homeostasis and Vascular Pathology. Arterioscler Thromb Vasc Biol 2021; 41:1607-1624. [PMID: 33691479 PMCID: PMC8057528 DOI: 10.1161/atvbaha.120.315697] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/01/2021] [Indexed: 12/12/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Anton G. Kutikhin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Lian Feenstra
- Department of Pathology and Medical Biology, Division of Pathology (L.F., J.-L.H.), University Medical Center Groningen, University of Groningen, the Netherlands
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology (L.F., G.K.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Alexander E. Kostyunin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Arseniy E. Yuzhalin
- Laboratory for Vascular Biology, Division of Experimental and Clinical Cardiology, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo, Russian Federation (A.G.K., A.E.K., A.E.Y.)
| | - Jan-Luuk Hillebrands
- Department of Pathology and Medical Biology, Division of Pathology (L.F., J.-L.H.), University Medical Center Groningen, University of Groningen, the Netherlands
| | - Guido Krenning
- Laboratory for Cardiovascular Regenerative Medicine, Department of Pathology and Medical Biology (L.F., G.K.), University Medical Center Groningen, University of Groningen, the Netherlands
- Sulfateq B.V., Admiraal de Ruyterlaan 5, 9726 GN, Groningen, the Netherlands (G.K.)
| |
Collapse
|
157
|
Deligiorgi MV, Panayiotidis MI, Siasos G, Trafalis DT. Osteoporosis Entwined with Cardiovascular Disease: The Implication of Osteoprotegerin and the Example of Statins. Curr Med Chem 2021; 28:1443-1467. [PMID: 31971101 DOI: 10.2174/0929867327666200123151132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 12/19/2019] [Accepted: 12/24/2019] [Indexed: 11/22/2022]
Abstract
Beyond being epiphenomenon of shared epidemiological factors, the integration of Osteoporosis (OP) with Cardiovascular Disease (CVD) - termed "calcification paradox" - reflects a continuum of aberrant cardiometabolic status. The present review provides background knowledge on "calcification paradox", focusing on the endocrine aspect of vasculature orchestrated by the osteoblastic molecular fingerprint of vascular cells, acquired via imbalance among established modulators of mineralization. Osteoprotegerin (OPG), the well-established osteoprotective cytokine, has recently been shown to exert a vessel-modifying role. Prompted by this notion, the present review interrogates OPG as the potential missing link between OP and CVD. However, so far, the confirmation of this hypothesis is hindered by the equivocal role of OPG in CVD, being both proatherosclerotic and antiatherosclerotic. Further research is needed to illuminate whether OPG could be a biomarker of the "calcification paradox". Moreover, the present review brings into prominence the dual role of statins - cardioprotective and osteoprotective - as a potential illustration of the integration of CVD with OP. Considering that the statins-induced modulation of OPG is central to the statins-driven osteoprotective signalling, statins could be suggested as an illustration of the role of OPG in the bone/vessels crosstalk, if further studies consolidate the contribution of OPG to the cardioprotective role of statins. Another outstanding issue that merits further evaluation is the inconsistency of the osteoprotective role of statins. Further understanding of the varying bone-modifying role of statins, likely attributed to the unique profile of different classes of statins defined by distinct physicochemical characteristics, may yield tangible benefits for treating simultaneously OP and CVD.
Collapse
Affiliation(s)
- Maria V Deligiorgi
- Department of Pharmacology - Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias, 11527 Goudi, Athens, Greece
| | - Mihalis I Panayiotidis
- Department of Applied Sciences, Group of Translational Biosciences, Faculty of Health & Life Sciences, Northumbria University, Ellison Building A516, Newcastle Upon Tyne, NE1 8ST, United Kingdom
| | - Gerasimos Siasos
- Department of Cardiology, Faculty of Medicine, 1st Hippokration Hospital, National and Kapodistrian University of Athens, 114 Vas Sofias, 11527 Athens, Greece
| | - Dimitrios T Trafalis
- Department of Pharmacology - Clinical Pharmacology Unit, Faculty of Medicine, National and Kapodistrian University of Athens, Building 16, 1st Floor, 75 Mikras Asias, 11527 Goudi, Athens, Greece
| |
Collapse
|
158
|
Liu W, Li CJ, Li LJ. [Advances in molecular mechanisms of bone invasion by oral cancer]. HUA XI KOU QIANG YI XUE ZA ZHI = HUAXI KOUQIANG YIXUE ZAZHI = WEST CHINA JOURNAL OF STOMATOLOGY 2021; 39:221-226. [PMID: 33834679 DOI: 10.7518/hxkq.2021.02.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Bone invasion by oral cancer is a common clinical problem, which affects the choice of treatment and predicts a poor prognosis. Unfortunately, the molecular mechanism of this phenomenon has not been fully elucidated. Current studies have revealed that oral cancer cells modulate the formation and function of osteoclasts through the expression of a series of signal molecules. Many signal pathways are involved in this process, of which receptor activator of nuclear factor-κB ligand/receptor activator of nuclear factor-κB/osteoprotegerin signaling pathway attracted much attention. In this review, we introduce recent progress in molecular mechanisms of bone invasion by oral cancer.
Collapse
Affiliation(s)
- Wei Liu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Chun-Jie Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Long-Jiang Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral Diseases & Dept. of Head and Neck Oncology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
159
|
Kim W, Tokuda H, Tanabe K, Yamaguchi S, Hioki T, Tachi J, Matsushima-Nishiwaki R, Kozawa O, Iida H. Acetaminophen reduces osteoprotegerin synthesis stimulated by PGE 2 and PGF 2α in osteoblasts: attenuation of SAPK/JNK but not p38 MAPK or p44/p42 MAPK. Biomed Res 2021; 42:77-84. [PMID: 33840687 DOI: 10.2220/biomedres.42.77] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Acetaminophen is one of the most widely used analgesic and antipyretic medicines, whose long-period use has reportedly been associated with an increased risk of bone fracture. However, the mechanism underlying this undesired effect remains to be investigated. The homeostatic control of bone tissue depends on the interaction between osteoblasts and osteoclasts. Osteoprotegerin produced by osteoblasts is known to play an essential role in suppressing osteoclast induction. We have previously reported that prostaglandin (PG) E2 and PGF2α induce osteoprotegerin synthesis through p38 mitogen-activated protein kinase (MAPK), p44/p42 MAPK and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in osteoblast-like MC3T3-E1 cells. In the present study, we investigated the effects of acetaminophen on the osteoprotegerin synthesis induced by PGE2 and PGF2α in MC3T3-E1 cells. Acetaminophen significantly suppressed the osteoprotegerin release stimulated by PGE2 and PGF2α. The PGE2-induced expression of osteoprotegerin mRNA was also reduced by acetaminophen. Acetaminophen markedly downregulated the phosphorylation of SAPK/JNK stimulated by PGE2 and PGF2α, but not those of p38 MAPK or p44/p42 MAPK. SP600125, an inhibitor of SAPK/JNK, suppressed the levels of PGE2- and PGF2α-upregulated osteoprotegerin mRNA expression. Taken together, these results strongly suggest that acetaminophen reduces the PGE2- and PGF2α-stimulated synthesis of osteoprotegerin in osteoblasts, and that the suppressive effect is exerted via attenuation of SAPK/JNK. These findings provide a molecular basis for the possible effect of acetaminophen on bone tissue metabolism.
Collapse
Affiliation(s)
- Woo Kim
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine.,Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine.,Department of Clinical Laboratory/Medical Genome Center, National Center for Geriatrics and Gerontology
| | - Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Shinobu Yamaguchi
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| | - Tomoyuki Hioki
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine.,Department of Dermatology, Kizawa Memorial Hospital
| | - Junko Tachi
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine.,Department of Pharmacology, Gifu University Graduate School of Medicine
| | | | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine
| | - Hiroki Iida
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine
| |
Collapse
|
160
|
Carrillo-López N, Martínez-Arias L, Fernández-Villabrille S, Ruiz-Torres MP, Dusso A, Cannata-Andía JB, Naves-Díaz M, Panizo S. Role of the RANK/RANKL/OPG and Wnt/β-Catenin Systems in CKD Bone and Cardiovascular Disorders. Calcif Tissue Int 2021; 108:439-451. [PMID: 33586001 DOI: 10.1007/s00223-020-00803-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 12/19/2020] [Indexed: 12/23/2022]
Abstract
In the course of chronic kidney disease (CKD), alterations in the bone-vascular axis augment the risk of bone loss, fractures, vascular and soft tissue calcification, left ventricular hypertrophy, renal and myocardial fibrosis, which markedly increase morbidity and mortality rates. A major challenge to improve skeletal and cardiovascular outcomes in CKD patients requires a better understanding of the increasing complex interactions among the main modulators of the bone-vascular axis. Serum parathyroid hormone (PTH), phosphorus (P), calcium (Ca), fibroblast growth factor 23 (FGF23), calcidiol, calcitriol and Klotho are involved in this axis interact with RANK/RANKL/OPG system and the Wnt/β-catenin pathway. The RANK/RANKL/OPG system controls bone remodeling by inducing osteoblast synthesis of RANKL and downregulating OPG production and it is also implicated in vascular calcification. The complexity of this system has recently increased due the discovery of LGR4, a novel RANKL receptor involved in bone formation, but possibly also in vascular calcification. The Wnt/β-catenin pathway plays a key role in bone formation: when this pathway is activated, bone is formed, but when it is inhibited, bone formation is stopped. In the progression of CKD, a downregulation of the Wnt/β-catenin pathway has been described which occurs mainly through the not coincident elevations of sclerostin, Dickkopf1 (Dkk1) and the secreted Frizzled Related Proteins (sFRPs). This review analyzes the interactions of PTH, P, Ca, FGF23, calcidiol, calcitriol and Klotho with the RANKL/RANKL/OPG system and the Wnt/β-catenin, pathway and their implications in bone and cardiovascular disorders in CKD.
Collapse
Affiliation(s)
- Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Sara Fernández-Villabrille
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - María Piedad Ruiz-Torres
- Department of System Biology, Universidad de Alcalá, Retic REDinREN-ISCIII, Alcalá de Henares, Spain
| | - Adriana Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain.
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Universidad de Oviedo, Retic REDinREN-ISCIII, Avda. Roma, sn., 33011, Oviedo, Spain.
| | | |
Collapse
|
161
|
Carrillo-López N, Martínez-Arias L, Alonso-Montes C, Martín-Carro B, Martín-Vírgala J, Ruiz-Ortega M, Fernández-Martín JL, Dusso AS, Rodriguez-García M, Naves-Díaz M, Cannata-Andía JB, Panizo S. The receptor activator of nuclear factor κΒ ligand receptor leucine-rich repeat-containing G-protein-coupled receptor 4 contributes to parathyroid hormone-induced vascular calcification. Nephrol Dial Transplant 2021; 36:618-631. [PMID: 33367746 DOI: 10.1093/ndt/gfaa290] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Accepted: 08/12/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND In chronic kidney disease, serum phosphorus (P) elevations stimulate parathyroid hormone (PTH) production, causing severe alterations in the bone-vasculature axis. PTH is the main regulator of the receptor activator of nuclear factor κB (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) system, which is essential for bone maintenance and also plays an important role in vascular smooth muscle cell (VSMC) calcification. The discovery of a new RANKL receptor, leucine-rich repeat-containing G-protein-coupled receptor 4 (LGR4), which is important for osteoblast differentiation but with an unknown role in vascular calcification (VC), led us to examine the contribution of LGR4 in high P/high PTH-driven VC. METHODS In vivo studies were conducted in subtotally nephrectomized rats fed a normal or high P diet, with and without parathyroidectomy (PTX). PTX rats were supplemented with PTH(1-34) to achieve physiological serum PTH levels. In vitro studies were performed in rat aortic VSMCs cultured in control medium, calcifying medium (CM) or CM plus 10-7 versus 10-9 M PTH. RESULTS Rats fed a high P diet had a significantly increased aortic calcium (Ca) content. Similarly, Ca deposition was higher in VSMCs exposed to CM. Both conditions were associated with increased RANKL and LGR4 and decreased OPG aorta expression and were exacerbated by high PTH. Silencing of LGR4 or parathyroid hormone receptor 1 (PTH1R) attenuated the high PTH-driven increases in Ca deposition. Furthermore, PTH1R silencing and pharmacological inhibition of protein kinase A (PKA), but not protein kinase C, prevented the increases in RANKL and LGR4 and decreased OPG. Treatment with PKA agonist corroborated that LGR4 regulation is a PTH/PKA-driven process. CONCLUSIONS High PTH increases LGR4 and RANKL and decreases OPG expression in the aorta, thereby favouring VC. The hormone's direct pro-calcifying actions involve PTH1R binding and PKA activation.
Collapse
Affiliation(s)
- Natalia Carrillo-López
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Laura Martínez-Arias
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Cristina Alonso-Montes
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Beatriz Martín-Carro
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Julia Martín-Vírgala
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Marta Ruiz-Ortega
- Vascular and Renal Laboratory Fundación Jimenez Díaz, Universidad Autónoma Madrid, REDinREN-ISCIII, Madrid, Spain
| | - José Luis Fernández-Martín
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Adriana S Dusso
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Minerva Rodriguez-García
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain.,Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Manuel Naves-Díaz
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| | - Jorge B Cannata-Andía
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain.,Departamento de Medicina, Universidad de Oviedo, Oviedo, Spain
| | - Sara Panizo
- Bone and Mineral Research Unit, Hospital Universitario Central de Asturias, Instituto de Investigación Sanitaria del Principado de Asturias, REDinREN-ISCIII, Oviedo, Spain
| |
Collapse
|
162
|
Shams R, Drasites KP, Zaman V, Matzelle D, Shields DC, Garner DP, Sole CJ, Haque A, Banik NL. The Pathophysiology of Osteoporosis after Spinal Cord Injury. Int J Mol Sci 2021; 22:3057. [PMID: 33802713 PMCID: PMC8002377 DOI: 10.3390/ijms22063057] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 03/15/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Spinal cord injury (SCI) affects approximately 300,000 people in the United States. Most individuals who sustain severe SCI also develop subsequent osteoporosis. However, beyond immobilization-related lack of long bone loading, multiple mechanisms of SCI-related bone density loss are incompletely understood. Recent findings suggest neuronal impairment and disability may lead to an upregulation of receptor activator of nuclear factor-κB ligand (RANKL), which promotes bone resorption. Disruption of Wnt signaling and dysregulation of RANKL may also contribute to the pathogenesis of SCI-related osteoporosis. Estrogenic effects may protect bones from resorption by decreasing the upregulation of RANKL. This review will discuss the current proposed physiological and cellular mechanisms explaining osteoporosis associated with SCI. In addition, we will discuss emerging pharmacological and physiological treatment strategies, including the promising effects of estrogen on cellular protection.
Collapse
Affiliation(s)
- Ramsha Shams
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Kelsey P. Drasites
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Vandana Zaman
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| | - Denise Matzelle
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| | - Donald C. Shields
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
| | - Dena P. Garner
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Christopher J. Sole
- Department of Health and Human Performance, The Citadel, 171 Moultrie St., Charleston, SC 29409, USA; (D.P.G.); (C.J.S.)
| | - Azizul Haque
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
| | - Narendra L. Banik
- Department of Neurosurgery, Medical University of South Carolina, 96 Jonathan Lucas St., Charleston, SC 29425, USA; (R.S.); (K.P.D.); (V.Z.); (D.M.); (D.C.S.)
- Department of Microbiology and Immunology, Medical University of South Carolina, 173 Ashley Avenue, Charleston, SC 29425, USA
- Ralph H. Johnson Veterans Administration Medical Center, 109 Bee St., Charleston, SC 29401, USA
| |
Collapse
|
163
|
Hathaway-Schrader JD, Novince CM. Maintaining homeostatic control of periodontal bone tissue. Periodontol 2000 2021; 86:157-187. [PMID: 33690918 DOI: 10.1111/prd.12368] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alveolar bone is a unique osseous tissue due to the proximity of dental plaque biofilms. Periodontal health and homeostasis are mediated by a balanced host immune response to these polymicrobial biofilms. Dysbiotic shifts within dental plaque biofilms can drive a proinflammatory immune response state in the periodontal epithelial and gingival connective tissues, which leads to paracrine signaling to subjacent bone cells. Sustained chronic periodontal inflammation disrupts "coupled" osteoclast-osteoblast actions, which ultimately result in alveolar bone destruction. This chapter will provide an overview of alveolar bone physiology and will highlight why the oral microbiota is a critical regulator of alveolar bone remodeling. The ecology of dental plaque biofilms will be discussed in the context that periodontitis is a polymicrobial disruption of host homeostasis. The pathogenesis of periodontal bone loss will be explained from both a historical and current perspective, providing the opportunity to revisit the role of fibrosis in alveolar bone destruction. Periodontal immune cell interactions with bone cells will be reviewed based on our current understanding of osteoimmunological mechanisms influencing alveolar bone remodeling. Lastly, probiotic and prebiotic interventions in the oral microbiota will be evaluated as potential noninvasive therapies to support alveolar bone homeostasis and prevent periodontal bone loss.
Collapse
Affiliation(s)
- Jessica D Hathaway-Schrader
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| | - Chad M Novince
- Department of Oral Health Sciences, College of Dental Medicine, Medical University of South Carolina, Charleston, South Carolina, USA
| |
Collapse
|
164
|
Yamaguchi M, Fukasawa S. Is Inflammation a Friend or Foe for Orthodontic Treatment?: Inflammation in Orthodontically Induced Inflammatory Root Resorption and Accelerating Tooth Movement. Int J Mol Sci 2021; 22:2388. [PMID: 33673606 PMCID: PMC7957544 DOI: 10.3390/ijms22052388] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
The aim of this paper is to provide a review on the role of inflammation in orthodontically induced inflammatory root resorption (OIIRR) and accelerating orthodontic tooth movement (AOTM) in orthodontic treatment. Orthodontic tooth movement (OTM) is stimulated by remodeling of the periodontal ligament (PDL) and alveolar bone. These remodeling activities and tooth displacement are involved in the occurrence of an inflammatory process in the periodontium, in response to orthodontic forces. Inflammatory mediators such as prostaglandins (PGs), interleukins (Ils; IL-1, -6, -17), the tumor necrosis factor (TNF)-α superfamily, and receptor activator of nuclear factor (RANK)/RANK ligand (RANKL)/osteoprotegerin (OPG) are increased in the PDL during OTM. OIIRR is one of the accidental symptoms, and inflammatory mediators have been detected in resorbed roots, PDL, and alveolar bone exposed to heavy orthodontic force. Therefore, these inflammatory mediators are involved with the occurrence of OIIRR during orthodontic tooth movement. On the contrary, regional accelerating phenomenon (RAP) occurs after fractures and surgery such as osteotomies or bone grafting, and bone healing is accelerated by increasing osteoclasts and osteoblasts. Recently, tooth movement after surgical procedures such as corticotomy, corticision, piezocision, and micro-osteoperforation might be accelerated by RAP, which increases the bone metabolism. Therefore, inflammation may be involved in accelerated OTM (AOTM). The knowledge of inflammation during orthodontic treatment could be used in preventing OIIRR and AOTM.
Collapse
Affiliation(s)
- Masaru Yamaguchi
- Ginza Orthodontic Clinic, Ginza Granvia 6F, 3-3-14 Ginza, Chuo-ku, Tokyo 104-0061, Japan;
| | | |
Collapse
|
165
|
Nishida D, Arai A, Zhao L, Yang M, Nakamichi Y, Horibe K, Hosoya A, Kobayashi Y, Udagawa N, Mizoguchi T. RANKL/OPG ratio regulates odontoclastogenesis in damaged dental pulp. Sci Rep 2021; 11:4575. [PMID: 33633362 PMCID: PMC7907144 DOI: 10.1038/s41598-021-84354-y] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Accepted: 02/16/2021] [Indexed: 01/31/2023] Open
Abstract
Bone-resorbing osteoclasts are regulated by the relative ratio of the differentiation factor, receptor activator NF-kappa B ligand (RANKL) and its decoy receptor, osteoprotegerin (OPG). Dental tissue-localized-resorbing cells called odontoclasts have regulatory factors considered as identical to those of osteoclasts; however, it is still unclear whether the RANKL/OPG ratio is a key factor for odontoclast regulation in dental pulp. Here, we showed that odontoclast regulators, macrophage colony-stimulating factor-1, RANKL, and OPG were detectable in mouse pulp of molars, but OPG was dominantly expressed. High OPG expression was expected to have a negative regulatory effect on odontoclastogenesis; however, odontoclasts were not detected in the dental pulp of OPG-deficient (KO) mice. In contrast, damage induced odontoclast-like cells were seen in wild-type pulp tissues, with their number significantly increased in OPG-KO mice. Relative ratio of RANKL/OPG in the damaged pulp was significantly higher than in undamaged control pulp. Pulp damages enhanced hypoxia inducible factor-1α and -2α, reported to increase RANKL or decrease OPG. These results reveal that the relative ratio of RANKL/OPG is significant to pulpal odontoclastogenesis, and that OPG expression is not required for maintenance of pulp homeostasis, but protects pulp from odontoclastogenesis caused by damages.
Collapse
Affiliation(s)
- Daisuke Nishida
- grid.265070.60000 0001 1092 3624Oral Health Science Center, Tokyo Dental College, Tokyo, 101-0061 Japan
| | - Atsushi Arai
- grid.411611.20000 0004 0372 3845Department of Orthodontics, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Lijuan Zhao
- grid.411611.20000 0004 0372 3845Institute for Oral Science, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Mengyu Yang
- grid.411611.20000 0004 0372 3845Institute for Oral Science, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Yuko Nakamichi
- grid.411611.20000 0004 0372 3845Institute for Oral Science, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Kanji Horibe
- grid.411611.20000 0004 0372 3845Department of Oral Histology, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Akihiro Hosoya
- grid.412021.40000 0004 1769 5590Department of Histology, School of Dentistry, Health Sciences University of Hokkaido, Hokkaido, 061-0293 Japan
| | - Yasuhiro Kobayashi
- grid.411611.20000 0004 0372 3845Institute for Oral Science, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Nobuyuki Udagawa
- grid.411611.20000 0004 0372 3845Department of Oral Biochemistry, Matsumoto Dental University, Nagano, 399-0781 Japan
| | - Toshihide Mizoguchi
- grid.265070.60000 0001 1092 3624Oral Health Science Center, Tokyo Dental College, Tokyo, 101-0061 Japan ,grid.411611.20000 0004 0372 3845Department of Oral Biochemistry, Matsumoto Dental University, Nagano, 399-0781 Japan
| |
Collapse
|
166
|
Lin S, Li X, Zhang J, Zhang Y. Omentin-1: Protective impact on ischemic stroke via ameliorating atherosclerosis. Clin Chim Acta 2021; 517:31-40. [PMID: 33607071 DOI: 10.1016/j.cca.2021.02.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 02/03/2021] [Accepted: 02/05/2021] [Indexed: 12/26/2022]
Abstract
Omentin-1, a newly identified adipokine, has recently been revealed as a novel biomarker for ischemic stroke (IS). Low circulating omentin-1 levels could indicate a high risk of IS, and elevated omentin-1 levels exert a favorable impact on cerebral ischemia. Furthermore, omentin-1 has anti-atherosclerotic, anti-inflammatory, and cardiovascular protective capabilities through the intracellular Akt/AMP-activated protein kinase (AMPK)/ nuclear factor-κB (NF-κB) and certain protein kinase (ERK, JNK, and p38) signaling pathways. Omentin-1 also alleviates endothelial cell dysfunction, improves revascularization via the Akt-endothelial nitric-oxide synthase (eNOS) regulatory axis, promotes endothelium-dependent vasodilation through endothelium-derived NO in an eNOS fashion, and inhibits VSMC proliferation by means of AMPK/ERK signaling pathways, VSMC migration via inactivation of the NADPH oxidase (NOX)/ROS/p38/HSP27 pathways and artery calcification via the PI3K-Akt pathway. These findings indicate that omentin-1 may be a negative mediator of IS. Pharmacologically, several lines of clinical evidence indicate that metformin and statins could elevate omentin-1 levels, although the specific mechanism has not been precisely delineated until now. This study is the first to summarize the comprehensive mechanisms between omentin-1 and atherosclerosis and to review the shielding effect of omentin-1 on IS. We shed light on omentin-1 as a novel therapeutic target for combating IS.
Collapse
Affiliation(s)
- Shiyi Lin
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China; School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China; School of Pharmacy, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Xin Li
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China
| | - Jiabei Zhang
- School of Life Sciences, Westlake University, Hangzhou 310024, China; Institute of Biology, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Yuyang Zhang
- School of Life Science and Biopharmaceutics, Shenyang Pharmaceutical University, Shenyang 110016, China.
| |
Collapse
|
167
|
Mohammadi B, Esmaeilizade Z, Omrani MD, Ghaderian SMH, Rajabibazl M, Fazeli Z. The Effect of Co-treating Human Mesenchymal Stem Cells with Epigallocatechin Gallate and Hypoxia-Inducible Factor-1 on the Expression of RANKL/RANK/OPG Signaling Pathway, Osteogenesis, and Angiogenesis Genes. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2021. [DOI: 10.1007/s40883-021-00197-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
168
|
Jung S, Joo NS, Kim YN, Choi BH. Cut-off value of serum homocysteine in relation to increase of coronary artery calcification. J Investig Med 2021; 69:345-350. [PMID: 33148632 PMCID: PMC7848052 DOI: 10.1136/jim-2020-001478] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2020] [Indexed: 01/07/2023]
Abstract
A recent study reported that coronary artery calcification (CAC) and serum homocysteine were well associated; however, no report is available for the cut-off value of serum homocysteine according to increase of coronary-artery calcification volume score (CVS). The data of 469 out of 777 subjects in 1 health promotion center located in Seoul were selected after exclusion of the missing data of serum homocysteine and CVS. CVS was categorized into 2 groups: CVS=0 and CVS>0. Serum homocysteine according to the CVS groups was compared, and the cut-off value of serum homocysteine according to the increase of CVS (>0) was calculated using the receiver operating characteristic curve. Mean age was 54.5 years and the proportion of females was 22.2%. Mean serum homocysteine concentration and CVS were 11.2 μmol/L and 50.4, respectively. After adjustments for age and sex, serum homocysteine was associated with CVS (r=0.167, p=0.001), and Log(Homocysteine) also showed a significant difference according to the CVS groups. The cut-off value of serum homocysteine according to the increase of CVS (>0) was 9.45 μmol/L (area under the curve=0.569 (95% CI 0.512 to 0.625), p=0.015). The cut-off value of serum homocysteine was 9.45 μmol/L according to the increase of coronary-artery CVS.
Collapse
Affiliation(s)
- Susie Jung
- Department of Family Practice and Community Health, Ajou University School of Medicine, Suwon, South Korea
| | - Nam-Seok Joo
- Department of Family Practice and Community Health, Ajou University Hospital, Suwon, Gyeonggi-do, South Korea
| | - Yu-Na Kim
- Department of Family Practice and Community Health, Ajou University School of Medicine, Suwon, South Korea
| | - Beom-Hee Choi
- Functional Medicine Clinic, GCIMED, Seoul, South Korea
| |
Collapse
|
169
|
Salam S, Gallagher O, Gossiel F, Paggiosi M, Eastell R, Khwaja A. Vascular calcification relationship to vascular biomarkers and bone metabolism in advanced chronic kidney disease. Bone 2021; 143:115699. [PMID: 33091638 DOI: 10.1016/j.bone.2020.115699] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 10/09/2020] [Accepted: 10/15/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Vascular calcification (VC) and renal osteodystrophy are important complications of advanced chronic kidney disease (CKD). High resolution peripheral quantitative computed tomography (HRpQCT) is able to assess bone microstructure in renal osteodystrophy and lower leg arterial calcification (LLAC) is usually seen as an incidental finding. LLAC can be a useful quantitative assessment of VC in CKD but the relationship between LLAC and vascular biomarkers and bone is unknown. We aimed to assess the relationship between LLAC and biomarkers, bone turnover and microstructure. METHODS In this cross-sectional study, fasting blood samples were taken from 69 CKD stages 4-5D patients and 68 healthy controls. HRpQCT of distal tibia and radius were performed. 43 CKD patients had trans-iliac bone biopsy after tetracycline labelling. RESULTS LLAC was more severe in CKD than controls (median [IQR] 1.043 [0.05-16.52] vs 0 [0-0.55] mgHA, p < 0.001). CKD patients with diabetes (28%) had significantly higher LLAC compared to non-diabetic CKD (median [IQR] 24.07 [3.42-61.30] vs 0.23 [0-3.78] mgHA, p < 0.001). LLAC mass in CKD correlated with serum phosphate (rho = 0.29, p < 0.05), calcium x phosphate product (rho = 0.31, p < 0.05), intact parathyroid hormone (rho = 0.38, p < 0.01), intact fibroblast growth factor-23 (iFGF23) (rho = 0.40, p = 0.001), total alkaline phosphatase (rho = 0.41, p < 0.001), bone alkaline phosphatase (rho = 0.29, p < 0.05), osteocalcin (rho = 0.32, p < 0.05), osteoprotegerin (rho = 0.40, p = 0.001) and dephosphorylated-uncarboxylated matrix Gla protein (rho = 0.31, p < 0.05). LLAC in CKD also correlated with worse distal tibia cortical bone mineral density, thickness and porosity. No association was found between LLAC and bone turnover, mineralization or volume on biopsy in CKD. In multivariate analysis, only age, diabetes, iPTH and iFGF23 were independently associated with LLAC in CKD. CONCLUSIONS High levels of PTH and FGF23, along with older age and the presence of diabetes may all play independent roles in the development of LLAC in advanced CKD.
Collapse
Affiliation(s)
- Syazrah Salam
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, United Kingdom; Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom.
| | - Orla Gallagher
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Fatma Gossiel
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Margaret Paggiosi
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Richard Eastell
- Academic Unit of Bone Metabolism and Mellanby Centre for Bone Research, University of Sheffield, United Kingdom
| | - Arif Khwaja
- Sheffield Kidney Institute, Sheffield Teaching Hospitals NHS Foundation Trust, United Kingdom
| |
Collapse
|
170
|
Honma M, Ikebuchi Y, Suzuki H. RANKL as a key figure in bridging between the bone and immune system: Its physiological functions and potential as a pharmacological target. Pharmacol Ther 2021; 218:107682. [DOI: 10.1016/j.pharmthera.2020.107682] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/09/2020] [Indexed: 02/06/2023]
|
171
|
Kanji S, Sarkar R, Pramanik A, Kshirsagar S, Greene CJ, Das H. Dental pulp-derived stem cells inhibit osteoclast differentiation by secreting osteoprotegerin and deactivating AKT signalling in myeloid cells. J Cell Mol Med 2021; 25:2390-2403. [PMID: 33511706 PMCID: PMC7933945 DOI: 10.1111/jcmm.16071] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Osteoclasts (OCs) differentiate from the monocyte/macrophage lineage, critically regulate bone resorption and remodelling in both homeostasis and pathology. Various immune and non‐immune cells help initiating activation of myeloid cells for differentiation, whereas hyper‐activation leads to pathogenesis, and mechanisms are yet to be completely understood. Herein, we show the efficacy of dental pulp–derived stem cells (DPSCs) in limiting RAW 264.7 cell differentiation and underlying molecular mechanism, which has the potential for future therapeutic application in bone‐related disorders. We found that DPSCs inhibit induced OC differentiation of RAW 264.7 cells when co‐cultured in a contact‐free system. DPSCs reduced expression of key OC markers, such as NFATc1, cathepsin K, TRAP, RANK and MMP‐9 assessed by quantitative RT‐PCR, Western blotting and immunofluorescence detection methods. Furthermore, quantitative RT‐PCR analysis revealed that DPSCs mediated M2 polarization of RAW 264.7 cells. To define molecular mechanisms, we found that osteoprotegerin (OPG), an OC inhibitory factor, was up‐regulated in RAW 264.7 cells in the presence of DPSCs. Moreover, DPSCs also constitutively secrete OPG that contributed in limiting OC differentiation. Finally, the addition of recombinant OPG inhibited OC differentiation in a dose‐dependent manner by reducing the expression of OC differentiation markers, NFATc1, cathepsin K, TRAP, RANK and MMP9 in RAW 264.7 cells. RNAKL and M‐CSF phosphorylate AKT and activate PI3K‐AKT signalling pathway during osteoclast differentiation. We further confirmed that OPG‐mediated inhibition of the downstream activation of PI3K‐AKT signalling pathway was similar to the DPSC co‐culture–mediated inhibition of OC differentiation. This study provides novel evidence of DPSC‐mediated inhibition of osteoclastogenesis mechanisms.
Collapse
Affiliation(s)
- Suman Kanji
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Ripon Sarkar
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Asmita Pramanik
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Sudhir Kshirsagar
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Carl J Greene
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| | - Hiranmoy Das
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, TX, USA
| |
Collapse
|
172
|
Zhang Q, Chen T, Zhang Y, Lyu L, Zhang B, Huang C, Zhou X, Wu Y, Li Z. MiR-30c-5p regulates adventitial progenitor cells differentiation to vascular smooth muscle cells through targeting OPG. Stem Cell Res Ther 2021; 12:67. [PMID: 33468212 PMCID: PMC7814722 DOI: 10.1186/s13287-020-02127-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 12/28/2020] [Indexed: 11/28/2022] Open
Abstract
Background As the most important component of the vascular wall, vascular smooth muscle cells (VSMCs) participate in the pathological process by phenotype transformation or differentiation from stem/progenitor cells. The main purpose of this study was to reveal the role and related molecular mechanism of microRNA-30c-5p (miR-30c-5p) in VSMC differentiation from adventitial progenitor cells expressing stem cell antigen-1(Sca-1). Methods In this study, we detected the expression of miR-30c-5p in human normal peripheral arteries and atherosclerotic arteries. In vitro, a stable differentiation model from adventitial Sca-1+ progenitor cells to VSMCs was established using transforming growth factor-β1 (TGF-β1) induction and the expression of miR-30c-5p during the process was observed. Then, we explored the effect of miR-30c-5p overexpression and inhibition on the differentiation from adventitial Sca-1+ progenitor cells to VSMCs. The target genes of miR-30c-5p were identified by protein chip and biological analyses and the expression of these genes in the differentiation process were detected. Further, the relationship between the target gene and miR-30c-5p and its effect on differentiation were evaluated. Finally, the co-transfection of miR-30c-5p inhibitor and small interfering RNA (siRNA) of the target gene was implemented to verify the functional target gene of miR-30c-5p during the differentiation from adventitial Sca-1+ progenitor cells to VSMCs, and the dual-luciferase reporter gene assay was performed to detect whether the mRNA 3′untranslated region (UTR) of the target gene is the direct binding site of miR-30c-5p. Results The expression of miR-30c-5p in the human atherosclerotic arteries was significantly lower than that in the normal arteries. During the differentiation from adventitial Sca-1+ progenitor cells to VSMCs, the expression of VSMC special markers including smooth muscle α-actin (SMαA), smooth muscle-22α (SM22α), smooth muscle myosin heavy chain (SMMHC), and h1-caponin increased accompanied with cell morphology changing from elliptic to fusiform. Meanwhile, the expression of miR-30c-5p decreased significantly. In functional experiments, overexpression of miR-30c-5p inhibited SMαA, SM22α, SMMHC, and h1-caponin at the mRNA and protein levels. In contrast, inhibition of miR-30c-5p promoted the expression of SMαA, SM22α, SMMHC, and h1-caponin. The target gene, osteoprotegerin (OPG), was predicted through protein chip and bioinformatics analyses. Overexpression of miR-30c-5p inhibited OPG expression while inhibition of miR-30c-5p had an opposite effect. Co-transfection experiments showed that low expression of OPG could weaken the promotion effect of miR-30c-5p inhibitor on the differentiation from adventitial Sca-1+ progenitor cells to VSMCs and the dual-luciferase reporter gene assay demonstrated that miR-30c-5p could target the mRNA 3′UTR of OPG directly. Conclusions This study demonstrates that miR-30c-5p expression was significantly decreased in atherosclerotic arteries and miR-30c-5p inhibited VSMC differentiation from adventitial Sca-1+ progenitor cells through targeting OPG, which may provide a new target for the treatment of VSMCs-associated diseases.
Collapse
Affiliation(s)
- Qing Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Ting Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Yun Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Lingxia Lyu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Bohuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Chengchen Huang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Xuhao Zhou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China
| | - Yutao Wu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China.
| | - Zhoubin Li
- Department of Lung Transplantation, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang Province, P.R. China.
| |
Collapse
|
173
|
Role of Uremic Toxins in Early Vascular Ageing and Calcification. Toxins (Basel) 2021; 13:toxins13010026. [PMID: 33401534 PMCID: PMC7824162 DOI: 10.3390/toxins13010026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
In patients with advanced chronic kidney disease (CKD), the accumulation of uremic toxins, caused by a combination of decreased excretion secondary to reduced kidney function and increased generation secondary to aberrant expression of metabolite genes, interferes with different biological functions of cells and organs, contributing to a state of chronic inflammation and other adverse biologic effects that may cause tissue damage. Several uremic toxins have been implicated in severe vascular smooth muscle cells (VSMCs) changes and other alterations leading to vascular calcification (VC) and early vascular ageing (EVA). The above mentioned are predominant clinical features of patients with CKD, contributing to their exceptionally high cardiovascular mortality. Herein, we present an update on pathophysiological processes and mediators underlying VC and EVA induced by uremic toxins. Moreover, we discuss their clinical impact, and possible therapeutic targets aiming at preventing or ameliorating the harmful effects of uremic toxins on the vasculature.
Collapse
|
174
|
Li H, Xiao Z, Quarles LD, Li W. Osteoporosis: Mechanism, Molecular Target and Current Status on Drug Development. Curr Med Chem 2021; 28:1489-1507. [PMID: 32223730 PMCID: PMC7665836 DOI: 10.2174/0929867327666200330142432] [Citation(s) in RCA: 121] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/25/2020] [Accepted: 02/26/2020] [Indexed: 11/22/2022]
Abstract
CDATA[Osteoporosis is a pathological loss of bone mass due to an imbalance in bone remodeling where osteoclast-mediated bone resorption exceeds osteoblast-mediated bone formation resulting in skeletal fragility and fractures. Anti-resorptive agents, such as bisphosphonates and SERMs, and anabolic drugs that stimulate bone formation, including PTH analogues and sclerostin inhibitors, are current treatments for osteoporosis. Despite their efficacy, severe side effects and loss of potency may limit the long term usage of a single drug. Sequential and combinational use of current drugs, such as switching from an anabolic to an anti-resorptive agent, may provide an alternative approach. Moreover, there are novel drugs being developed against emerging new targets such as Cathepsin K and 17β-HSD2 that may have less side effects. This review will summarize the molecular mechanisms of osteoporosis, current drugs for osteoporosis treatment, and new drug development strategies.
Collapse
Affiliation(s)
- Hanxuan Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Zhousheng Xiao
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38165, USA
| | - L. Darryl Quarles
- Department of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38165, USA
| | - Wei Li
- Department of Pharmaceutical Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| |
Collapse
|
175
|
Udagawa N, Koide M, Nakamura M, Nakamichi Y, Yamashita T, Uehara S, Kobayashi Y, Furuya Y, Yasuda H, Fukuda C, Tsuda E. Osteoclast differentiation by RANKL and OPG signaling pathways. J Bone Miner Metab 2021; 39:19-26. [PMID: 33079279 DOI: 10.1007/s00774-020-01162-6] [Citation(s) in RCA: 373] [Impact Index Per Article: 93.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Accepted: 09/22/2020] [Indexed: 12/19/2022]
Abstract
INTRODUCTION In bone tissue, bone resorption by osteoclasts and bone formation by osteoblasts are repeated continuously. Osteoclasts are multinucleated cells that derive from monocyte-/macrophage-lineage cells and resorb bone. In contrast, osteoblasts mediate osteoclastogenesis by expressing receptor activator of nuclear factor-kappa B ligand (RANKL), which is expressed as a membrane-associated cytokine. Osteoprotegerin (OPG) is a soluble RANKL decoy receptor that is predominantly produced by osteoblasts and which prevents osteoclast formation and osteoclastic bone resorption by inhibiting the RANKL-RANKL receptor interaction. MATERIALS AND METHODS In this review, we would like to summarize our experimental results on signal transduction that regulates the expression of RANKL and OPG. RESULTS Using OPG gene-deficient mice, we have demonstrated that OPG and sclerostin produced by osteocytes play an important role in the maintenance of cortical and alveolar bone. In addition, it was shown that osteoclast-derived leukemia inhibitory factor (LIF) reduces the expression of sclerostin in osteocytes and promotes bone formation. WP9QY (W9) is a peptide that was designed to be structurally similar to one of the cysteine-rich TNF-receptortype-I domains. Addition of the W9 peptide to bone marrow culture simultaneously inhibited osteoclast differentiation and stimulated osteoblastic cell proliferation. An anti-sialic acid-binding immunoglobulin-like lectin 15 (Siglec-15) antibody inhibited multinucleated osteoclast formation induced by RANKL and macrophage colony-stimulating factor (M-CSF). Pit-forming activity of osteoclasts was also inhibited by the anti-Siglec-15 antibody. In addition, anti-Siglec-15 antibody treatment stimulated the appearance of osteoblasts in cultures of mouse bone marrow cells in the presence of RANKL and M-CSF. CONCLUSIONS Bone mass loss depends on the RANK-RANKL-OPG system, which is a major regulatory system of osteoclast differentiation induction, activation, and survival.
Collapse
Affiliation(s)
- Nobuyuki Udagawa
- Department of Biochemistry, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri, Nagano, 399-0781, Japan.
- Division of Hard Tissue Research, I, nstitute for Oral Science, Matsumoto Dental University, Nagano, 399-0781, Japan.
| | - Masanori Koide
- Division of Hard Tissue Research, I, nstitute for Oral Science, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Midori Nakamura
- Department of Biochemistry, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri, Nagano, 399-0781, Japan
- Division of Hard Tissue Research, I, nstitute for Oral Science, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Yuko Nakamichi
- Division of Hard Tissue Research, I, nstitute for Oral Science, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Teruhito Yamashita
- Division of Hard Tissue Research, I, nstitute for Oral Science, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Shunsuke Uehara
- Department of Biochemistry, Matsumoto Dental University, 1780 Gobara, Hiro-oka, Shiojiri, Nagano, 399-0781, Japan
| | - Yasuhiro Kobayashi
- Division of Hard Tissue Research, I, nstitute for Oral Science, Matsumoto Dental University, Nagano, 399-0781, Japan
| | - Yuriko Furuya
- Nagahama Institute for Biochemical Science, Oriental Yeast Co., Ltd., Nagahama, Japan
| | - Hisataka Yasuda
- Nagahama Institute for Biochemical Science, Oriental Yeast Co., Ltd., Nagahama, Japan
| | - Chie Fukuda
- Specialty Medicine Research Laboratories 1, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Eisuke Tsuda
- Specialty Medicine Research Laboratories 1, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
176
|
Qu H, Zhuang Y, Zhu L, Zhao Z, Wang K. The effects of vasoactive intestinal peptide on RANKL-induced osteoclast formation. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:127. [PMID: 33569429 PMCID: PMC7867916 DOI: 10.21037/atm-20-7607] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Congenital pseudarthrosis of the tibia is a rare disease characterized by an imbalance in bone remodeling. Vasoactive intestinal peptide (VIP) has been proven to modulate bone resorption and the formation of osteoclasts. This study aimed to explore the effects of VIP on the homeostasis of bone metabolism in diverse in vitro systems. Methods Bone marrow-derived macrophages (BMMs) were differentiated into tartrate-resistant acid phosphatase-positive cells through incubation with receptor activator of nuclear factor κB ligand (RANKL) and macrophage colony-stimulating factor (M-CSF). In vitro resorption pit detection was carried out to assess the effects of VIP on osteoclastic activity. Rat osteosarcoma cell line ROS 17/2.8 was cultured alone or co-cultured with rat BMMs in the presence or absence of VIP at various concentrations. The expression levels of RANKL, RANK, OPG, NF-κB, IL-6, ERK, CAII, and GAPDH were determined by qRT-PCR and WB assay. Results VIP was observed to repress osteoclast differentiation without affecting the number of osteoclast precursor cells. Furthermore, the modulation of the RANKL/osteoprotegerin (OPG), nuclear factor-κB (NF-κB), and extracellular signal-regulated kinase (ERK) signaling pathways were involved in the inhibitive influence of VIP upon bone erosion. Additionally, VIP affected the expression levels of osteoclastic factors including RANKL, OPG, and interleukin-6 in osteoblast cells. Furthermore, the expression levels of RANKL and RANK were increased, while OPG expression was reduced after treatment with VIP in the co-culture of ROS 17/2.8 and rat BMMs. ERK and NF-κB signal pathways were demonstrated to be involved in the effect of VIP in the co-culture system. Conclusions VIP plays a critical role in bone remodeling and might serve as a potential target in the development of treatments for congenital pseudarthrosis of the tibia.
Collapse
Affiliation(s)
- Hongyi Qu
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Department of Pediatric Surgery, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, China
| | - Yan Zhuang
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Lin Zhu
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Zuohui Zhao
- Department of Pediatric Surgery, The First Affiliated Hospital of Shandong First Medical University (Shandong Provincial Qianfoshan Hospital), Jinan, China
| | - Kelai Wang
- Department of Pediatric Surgery, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
177
|
Chen Y, Zhao X, Wu H. Transcriptional Programming in Arteriosclerotic Disease: A Multifaceted Function of the Runx2 (Runt-Related Transcription Factor 2). Arterioscler Thromb Vasc Biol 2021; 41:20-34. [PMID: 33115268 PMCID: PMC7770073 DOI: 10.1161/atvbaha.120.313791] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Despite successful therapeutic strategies in the prevention and treatment of arteriosclerosis, the cardiovascular complications remain a major clinical and societal issue worldwide. Increased vascular calcification promotes arterial stiffness and accelerates cardiovascular morbidity and mortality. Upregulation of the Runx2 (Runt-related transcription factor 2), an essential osteogenic transcription factor for bone formation, in the cardiovascular system has emerged as an important regulator for adverse cellular events that drive cardiovascular pathology. This review discusses the regulatory mechanisms that are critical for Runx2 expression and function and highlights the dynamic and complex cross talks of a wide variety of posttranslational modifications, including phosphorylation, acetylation, ubiquitination, and O-linked β-N-acetylglucosamine modification, in regulating Runx2 stability, cellular localization, and osteogenic transcriptional activity. How the activation of an array of signaling cascades by circulating and local microenvironmental factors upregulates Runx2 in vascular cells and promotes Runx2-mediated osteogenic transdifferentiation of vascular smooth muscle cells and expression of inflammatory cytokines that accelerate macrophage infiltration and vascular osteoclast formation is summarized. Furthermore, the increasing appreciation of a new role of Runx2 upregulation in promoting vascular smooth muscle cell phenotypic switch, and Runx2 modulated by O-linked β-N-acetylglucosamine modification and Runx2-dependent repression of smooth muscle cell-specific gene expression are discussed. Further exploring the regulation of this key osteogenic transcription factor and its new perspectives in the vasculature will provide novel insights into the transcriptional regulation of vascular smooth muscle cell phenotype switch, reprograming, and vascular inflammation that promote the pathogenesis of arteriosclerosis.
Collapse
Affiliation(s)
- Yabing Chen
- Department of Pathology, University of Alabama at Birmingham
- Research Department, Birmingham Veterans Affairs Medical Center, Birmingham, Alabama 35294
| | - Xinyang Zhao
- Department of Biochemistry, University of Alabama at Birmingham
| | - Hui Wu
- Department of Integrative Biomedical & Diagnostic Sciences, Oregon Health and Science University School of Dentistry, Portland, Oregon 97239
| |
Collapse
|
178
|
Raisi-Estabragh Z, Biasiolli L, Cooper J, Aung N, Fung K, Paiva JM, Sanghvi MM, Thomson RJ, Curtis E, Paccou J, Rayner JJ, Werys K, Puchta H, Thomas KE, Lee AM, Piechnik SK, Neubauer S, Munroe PB, Cooper C, Petersen SE, Harvey NC. Poor Bone Quality is Associated With Greater Arterial Stiffness: Insights From the UK Biobank. J Bone Miner Res 2021; 36:90-99. [PMID: 32964541 PMCID: PMC7613252 DOI: 10.1002/jbmr.4164] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 07/26/2020] [Accepted: 08/05/2020] [Indexed: 12/20/2022]
Abstract
Osteoporosis and ischemic heart disease (IHD) represent important public health problems. Existing research suggests an association between the two conditions beyond that attributable to shared risk factors, with a potentially causal relationship. In this study, we tested the association of bone speed of sound (SOS) from quantitative heel ultrasound with (i) measures of arterial compliance from cardiovascular magnetic resonance (aortic distensibility [AD]); (ii) finger photoplethysmography (arterial stiffness index [ASI]); and (iii) incident myocardial infarction and IHD mortality in the UK Biobank cohort. We considered the potential mediating effect of a range of blood biomarkers and cardiometabolic morbidities and evaluated differential relationships by sex, menopause status, smoking, diabetes, and obesity. Furthermore, we considered whether associations with arterial compliance explained association of SOS with ischemic cardiovascular outcomes. Higher SOS was associated with lower arterial compliance by both ASI and AD for both men and women. The relationship was most consistent with ASI, likely relating to larger sample size available for this variable (n = 159,542 versus n = 18,229). There was no clear evidence of differential relationship by menopause, smoking, diabetes, or body mass index (BMI). Blood biomarkers appeared important in mediating the association for both men and women, but with different directions of effect and did not fully explain the observed effects. In fully adjusted models, higher SOS was associated with significantly lower IHD mortality in men, but less robustly in women. The association of SOS with ASI did not explain this observation. In conclusion, our findings support a positive association between bone and vascular health with consistent patterns of association in men and women. The underlying mechanisms are complex and appear to vary by sex. © 2020 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)
- Zahra Raisi-Estabragh
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health National Health Service (NHS) Trust, London, UK
| | - Luca Biasiolli
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Jackie Cooper
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health National Health Service (NHS) Trust, London, UK
| | - Nay Aung
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health National Health Service (NHS) Trust, London, UK
| | - Kenneth Fung
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health National Health Service (NHS) Trust, London, UK
| | - José M Paiva
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
| | - Mihir M Sanghvi
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health National Health Service (NHS) Trust, London, UK
| | - Ross J Thomson
- Department of Cardiology, Royal Free Hospital, Royal Free London NHS Foundation Trust, London, UK
| | - Elizabeth Curtis
- Medical Research Council (MRC) Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
| | - Julien Paccou
- Rheumatology Department, Lille University Hospital, CHU Lille, MABlab ULR 4490, 59037 Lille, France
| | - Jennifer J Rayner
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Konrad Werys
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Henrike Puchta
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Katharine E Thomas
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Aaron M Lee
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
| | - Stefan K Piechnik
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Stefan Neubauer
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, National Institute for Health Research Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Patricia B Munroe
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
| | - Cyrus Cooper
- Medical Research Council (MRC) Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
- National Institute for Health Research (NIHR) Oxford Biomedical Research Centre, University of Oxford, Oxford, UK
| | - Steffen E Petersen
- William Harvey Research Institute, National Institute for Health Research (NIHR) Barts Biomedical Research Centre, Queen Mary University of London, Charterhouse Square, London, UK
- Barts Heart Centre, St Bartholomew's Hospital, Barts Health National Health Service (NHS) Trust, London, UK
| | - Nicholas C Harvey
- Medical Research Council (MRC) Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK
- National Institute for Health Research (NIHR) Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK
| |
Collapse
|
179
|
Abstract
Almost a quarter century has passed since discovery of receptor activator of NF-κB ligand (RANKL). This discovery had a major impact on identification of mechanisms regulating osteoclast differentiation and function, establishment of a research field bridging bone and the immune system (osteoimmunology), and development of a fully human anti-RANKL neutralizing antibody (denosumab). Denosumab is now clinically available for treatment of osteoporosis and cancer-induced bone diseases in the US, Europe and many other countries, including Japan. Denosumab is a so-called blockbuster drug, with sales of 5.0 billion US dollars in 2019. This is a real success story from bench to bedside. In this review, the pivotal roles of the RANKL/RANK/OPG system in osteoclast differentiation and function are shown. RANKL is a ligand required for osteoclast generation, RANK is the receptor for RANKL, and osteoprotegerin (OPG) is a decoy receptor for RANKL. The review covers recent results showing the importance of RANKL on osteoblasts in regulation of osteogenesis and the role of RANKL-RANK dual signaling in coupling of bone resorption and formation, including demonstration of RANKL reverse signaling that we had previously hypothesized. Possible applications of anti-RANKL antibody in treatment of cancer are also discussed.
Collapse
Affiliation(s)
- Hisataka Yasuda
- Nagahama Institute for Biochemical Science, Oriental Yeast Co., Ltd., 50, Kano-cho, Nagahama, Shiga, 526-0804, Japan.
| |
Collapse
|
180
|
Deng Z, Hu W, Ai H, Chen Y, Dong S. The Dramatic Role of IFN Family in Aberrant Inflammatory Osteolysis. Curr Gene Ther 2021; 21:112-129. [PMID: 33245272 DOI: 10.2174/1566523220666201127114845] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 11/11/2020] [Accepted: 11/12/2020] [Indexed: 11/22/2022]
Abstract
Skeletal system has been considered a highly dynamic system, in which bone-forming osteoblasts and bone-resorbing osteoclasts go through a continuous remodeling cycle to maintain homeostasis of bone matrix. It has been well acknowledged that interferons (IFNs), acting as a subgroup of cytokines, not only have crucial effects on regulating immunology but also could modulate the dynamic balance of bone matrix. In the light of different isoforms, IFNs have been divided into three major categories in terms of amino acid sequences, recognition of specific receptors and biological activities. Currently, type I IFNs consist of a multi-gene family with several subtypes, of which IFN-α exerts pro-osteoblastogenic effects to activate osteoblast differentiation and inhibits osteoclast fusion to maintain bone matrix integrity. Meanwhile, IFN-β suppresses osteoblast-mediated bone remodeling as well as exhibits inhibitory effects on osteoclast differentiation to attenuate bone resorption. Type II IFN constitutes the only type, IFN-γ, which exerts regulatory effects on osteoclastic bone resorption and osteoblastic bone formation by biphasic ways. Interestingly, type III IFNs are regarded as new members of IFN family composed of four members, including IFN-λ1 (IL-29), IFN-λ2 (IL-28A), IFN-λ3 (IL-28B) and IFN-λ4, which have been certified to participate in bone destruction. However, the direct regulatory mechanisms underlying how type III IFNs modulate the metabolic balance of bone matrix, remains poorly elucidated. In this review, we have summarized functions of IFN family during physiological and pathological conditions and described the mechanisms by which IFNs maintain bone matrix homeostasis via affecting the osteoclast-osteoblast crosstalk. In addition, the potential therapeutic effects of IFNs on inflammatory bone destruction diseases such as rheumatoid arthritis (RA), osteoarthritis (OA) and infectious bone diseases are also well displayed, which are based on the predominant role of IFNs in modulating the dynamic equilibrium of bone matrix.
Collapse
Affiliation(s)
- Zihan Deng
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Wenhui Hu
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Hongbo Ai
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Yueqi Chen
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Shiwu Dong
- Department of Biomedical Materials Science, Third Military Medical University (Army Medical University), Chongqing 400038, China
| |
Collapse
|
181
|
Chung J, Kim HL, Lee JP, Lim WH, Seo JB, Kim SH, Zo JH, Kim MA. Association of the Serum Osteoprotegerin Level With Target Organ Damage in Patients at High Risk of Coronary Artery Disease. Circ J 2020; 85:69-76. [PMID: 33250498 DOI: 10.1253/circj.cj-20-0675] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
BACKGROUND There is little data as to whether osteoprotegerin (OPG) is associated with target organ damage (TOD), so we evaluated the association in patients at high risk of coronary artery disease (CAD). METHODS AND RESULTS A total of 349 patients who underwent invasive coronary angiography (ICA) for suspected CAD were prospectively recruited. During the index admission, 6 TOD parameters were collected: extent of CAD, glomerular filtration rate (GFR), left ventricular mass index (LVMI), E/e', brachial-ankle pulse wave velocity (baPWV), and ankle-brachial index (ABI). Serum OPG levels were measured using enzyme-linked immunosorbent assay. The OPG level was significantly higher in patients with ≥1 TOD parameter than in those without (314±186 vs. 202±74 pg/mL, P<0.001). For each TOD parameter, the serum OPG level was significantly higher in patients with TOD than in those without (P<0.05 for each) except for ABI. In correlation analysis, OPG was significantly associated with GFR, LVMI, E/e', baPWV and ABI (P<0.05 for each). The OPG concentration increased proportionally with increasing TOD (P<0.001). Higher OPG concentrations (≥198 pg/mL) was significantly associated with the presence of TOD (odds ratio 3.22; 95% confidence interval 1.51-6.85; P=0.002) even after controlling for potential confounders. CONCLUSIONS Serum OPG level was significantly associated with a variety of TOD in patients undergoing ICA. OPG may be a useful marker for TOD and in the risk stratification of patients at high risk of CAD.
Collapse
Affiliation(s)
- Jaehoon Chung
- Division of Cardiology, Department of Internal Medicine, National Medical Center
| | - Hack-Lyoung Kim
- Division of Cardiology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| | - Jung Pyo Lee
- Division of Nephrology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| | - Woo-Hyun Lim
- Division of Cardiology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| | - Jae-Bin Seo
- Division of Cardiology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| | - Sang-Hyun Kim
- Division of Cardiology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| | - Joo-Hee Zo
- Division of Cardiology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| | - Myung-A Kim
- Division of Cardiology, Department of Internal Medicine, Seoul National University College of Medicine, Boramae Medical Center
| |
Collapse
|
182
|
Uto K, Yoshizawa S, Aoki C, Nishikawa T, Oda H. Inhibition of extracellular matrix integrity attenuates the early phase of aortic medial calcification in a rodent model. Atherosclerosis 2020; 319:10-20. [PMID: 33453491 DOI: 10.1016/j.atherosclerosis.2020.12.015] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 12/12/2020] [Accepted: 12/18/2020] [Indexed: 02/05/2023]
Abstract
BACKGROUND AND AIMS The mechanism of vascular calcification (VC) resembles that of bone metabolism, and a correlation has frequently been reported between calcification and vascular extracellular matrix (ECM) regulating its integrity; however, the detailed mechanisms remain unclear. In this study, we examined how the vascular ECM, especially collagen metabolism, is involved in the process of VC. METHODS VC was modeled using 5-week-old male Sprague-Dawley rats fed a diet containing warfarin and vitamin K1 (WVK). Additionally, β-aminopropionitrile (BAPN) was administered to inhibit lysyl oxidase (LOX), which is an enzyme that mediates collagen cross-linking. Harvested aortic samples were analyzed by staining with alizarin red (AR), immunohistochemistry (IHC), transmission electron microscopy (TEM), and ex vivo microcomputed tomography (μCT). RESULTS Rats fed WVK developed increasing numbers of aortic medial calcifications (AMCs) over time. TEM images indicated punctate calcification within collagen fibers in the early phase of AMC. AR staining of translucent samples revealed the distribution and severity of calcification, and these lesions were significantly decreased in the BAPN group. Three-dimensional reconstructed μCT images that allowed the quantification of calcified volumes revealed that BAPN significantly reduced the bulk of calcification. Moreover, IHC showed that both LOX and collagen I were present around the sites of AMC, and thus the IHC-positive area was reduced in the BAPN group compared to the WVK group. CONCLUSIONS The results indicated that inhibition of LOX by BAPN attenuated AMC, and that collagen metabolism plays a significant role in the early pathogenesis of VC.
Collapse
Affiliation(s)
- Kenta Uto
- Department of Pathology, Division of Experimental Pathology, Tokyo Women's Medical University, Tokyo, Japan.
| | - Saeko Yoshizawa
- Department of Pathology, Division of Experimental Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Chiharu Aoki
- Department of Pathology, Division of Experimental Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Toshio Nishikawa
- Department of Surgical Pathology, Tokyo Women's Medical University, Tokyo, Japan
| | - Hideaki Oda
- Department of Pathology, Division of Experimental Pathology, Tokyo Women's Medical University, Tokyo, Japan
| |
Collapse
|
183
|
Molecular Genetics and Modifier Genes in Pseudoxanthoma Elasticum, a Heritable Multisystem Ectopic Mineralization Disorder. J Invest Dermatol 2020; 141:1148-1156. [PMID: 33341249 DOI: 10.1016/j.jid.2020.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 10/19/2020] [Accepted: 10/23/2020] [Indexed: 01/08/2023]
Abstract
In the past two decades, there has been great progress in identifying the molecular basis and pathomechanistic details in pseudoxanthoma elasticum (PXE), a heritable multisystem ectopic mineralization disorder. Although the identification of pathogenic variants in ABCC6 has been critical for understanding the disease process, genetic modifiers have been disclosed that explain the phenotypic heterogeneity of PXE. Adding to the genetic complexity of PXE are PXE-like phenotypes caused by pathogenic variants in other ectopic mineralization-associated genes. This review summarizes the current knowledge of the genetics and candidate modifier genes in PXE, a multifactorial disease at the genome-environment interface.
Collapse
|
184
|
Denosumab Recovers Aortic Arch Calcification During Long-Term Hemodialysis. Kidney Int Rep 2020; 6:605-612. [PMID: 33732975 PMCID: PMC7938059 DOI: 10.1016/j.ekir.2020.12.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 11/21/2020] [Accepted: 12/08/2020] [Indexed: 12/15/2022] Open
Abstract
Introduction Aortic arch calcification (AoAC) is related closely to mortality risk in patients undergoing maintenance dialysis. Recent experimentally obtained data suggest that osteoprotegerin/receptor activator for nuclear factor κB ligand signal transmission plays a role in de novo chondrogenic transition of vascular cells leading to calcification that is unrelated to bone metabolism. This study investigated the long-term effects of denosumab, an osteoprotegerin mimic peptide, on AoAC. Methods This study examined 58 patients with an 8 year vintage of dialysis at 1 center for observational study during 2009 to 2020. Denosumab was administered to 28 patients every 6 months. Blood chemical data were used. AoAC proportions were measured using a simple but computed tomography–equivalent computer-based chest X-ray analysis (calcified pieces of areas around the aorta). Results Blood chemical data of the control and denosumab groups that did not differ at the start showed differences of mineral metabolism after 30 months of observation. Remarkably, the AoAC proportion increased from 29.4% to 46.25% in the control group but decreased significantly from 25.0% to 20.0% (P < 0.01) in the denosumab group. Denosumab effects on decalcification were not observed 12 months after initiation. Conclusion We conclude that long-term use of denosumab is effective to reverse or treat AoAC in patients undergoing hemodialysis.
Collapse
|
185
|
Kaur M, Nagpal M, Singh M. Osteoblast-n-Osteoclast: Making Headway to Osteoporosis Treatment. Curr Drug Targets 2020; 21:1640-1651. [DOI: 10.2174/1389450121666200731173522] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 07/02/2020] [Accepted: 07/15/2020] [Indexed: 12/18/2022]
Abstract
Background:
Bone is a dynamic tissue that continuously undergoes the modeling and remodeling
process to maintain its strength and firmness. Bone remodeling is determined by the functioning
of osteoblast and osteoclast cells. The imbalance between the functioning of osteoclast and osteoblast
cells leads to osteoporosis. Osteoporosis is divided into primary and secondary osteoporosis.
Generally, osteoporosis is diagnosed by measuring bone mineral density (BMD) and various osteoblast
and osteoclast cell markers.
Methods:
Relevant literature reports have been studied and data has been collected using various
search engines like google scholar, scihub, sciencedirect, pubmed, etc. A thorough understanding of
the mechanism of bone targeting strategies has been discussed and related literature has been studied
and compiled.
Results:
Bone remodeling process has been described in detail including various approaches for targeting
bone. Several bone targeting moieties have been stated in detail along with their mechanisms.
Targeting of osteoclasts and osteoblasts using various nanocarriers has been discussed in separate sections.
The toxicity issues or Biosafety related to the use of nanomaterials have been covered.
Conclusion:
The treatment of osteoporosis targets the inhibition of bone resorption and the use of
agents that promote bone mineralization to slow disease progression. Current osteoporosis therapy involves
the use of targeting moieties such as bisphosphonates and tetracyclines for targeting various
drugs. Nanotechnology has been used for targeting various drug molecules such as RANKLinhibitors,
parathyroid hormone analogues, estrogen agonists and antagonists, Wnt signaling enhancer
and calcitonin specifically to bone tissue (osteoclast and osteoblasts). So, a multicomponent treatment
strategy targeting both the bone cells will be more effective rather than targeting only osteoclasts and
it will be a potential area of research in bone targeting used to treat osteoporosis.
The first section of the review article covers various aspects of bone targeting. Another section comprises
details of various targeting moieties such as bisphosphonates, tetracyclines; and various
nanocarriers developed to target osteoclast and osteoblast cells and summarized data on in vivo models
has been used for assessment of bone targeting, drawbacks of current strategies and future perspectives.
Collapse
Affiliation(s)
- Malkiet Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Manjinder Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| |
Collapse
|
186
|
Harrak H, René E, Alsalemi N, Elftouh N, Lafrance JP. Osteoporotic fracture rates in chronic hemodialysis and effect of heparin exposure: a retrospective cohort study. BMC Nephrol 2020; 21:261. [PMID: 32646504 PMCID: PMC7350680 DOI: 10.1186/s12882-020-01916-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 06/28/2020] [Indexed: 11/12/2022] Open
Abstract
Background Patients receiving chronic hemodialysis treatments are at a higher risk of fracture compared to the general population. While the use of heparin during dialysis is crucial to avoid thrombosis of the extracorporeal circuit, the association of unfractionated heparin (UFH) and the risk of osteoporotic fracture has been shown for many years. However, this association was not as clear for low-molecular-weight heparin (LMWH) and the few collected data originated from studies among pregnant women. Our aim was to measure osteoporotic fracture rate among hemodialysis patients and to evaluate the association of LMWH compared to UFH in hemodialysis. Methods A retrospective cohort study was conducted on data extracted from the RAMQ and Med-Echo databases from January 2007 to March 2013 with patients chronically hemodialyzed in 21 participating centers. Incidence rates for each fracture sites were measured per 1000 patient-year (p-y) and their 95% confidence intervals (CI). Osteoporotic fracture risk for a first event with LMWH compared to UFH was estimated using a cox proportional hazard model using demographics, comorbidities and drug use as covariates. Results 4796 patients undergoing chronic hemodialysis were identified. The incidence rate for all fracture sites was 22.7 /1000 p-y (95% CI: 19.6–26.1) and 12.8 /1000 p-y (95% CI: 10.5–15.4) for hip and femur fractures. We found a similar risk of osteoporotic fracture for LMWH compared to UFH (adjusted HR = 1.01; 95%CI: 0.72–1.42). Age and malignancy increased the risk of fracture while cerebrovascular disease decreased the risk of fracture. Conclusions Compared to UFH, LMWH did not change the risk of osteoporotic fracture when used for the extracorporeal circuit anticoagulation in chronic hemodialysis.
Collapse
|
187
|
Mukai H, Dai L, Chen Z, Lindholm B, Ripsweden J, Brismar TB, Heimbürger O, Barany P, Qureshi AR, Söderberg M, Bäck M, Stenvinkel P. Inverse J-shaped relation between coronary arterial calcium density and mortality in advanced chronic kidney disease. Nephrol Dial Transplant 2020; 35:1202-1211. [PMID: 30534995 DOI: 10.1093/ndt/gfy352] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/28/2018] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND The coronary artery calcium (CAC) score from cardiac computed tomography (CT) is a composite of CAC volume and CAC density. In the general population, CAC volume is positively and CAC density inversely associated with cardiovascular disease (CVD) events, implying that decreased CAC density reflects atherosclerotic plaque instability. We analysed associations of CAC indices with mortality risk in patients with end-stage renal disease [chronic kidney disease Stage 5 (CKD5)]. METHODS In 296 CKD5 patients undergoing cardiac CT (median age 55 years, 67% male, 19% diabetes, 133 dialysed), the Framingham risk score (FRS), presence of CVD and protein-energy wasting (PEW; subjective global assessment) and high-sensitivity C-reactive protein (hsCRP) and interleukin-6 (IL-6) were determined at baseline. During follow-up for a median of 35 months, 51 patients died and 75 patients underwent renal transplantation. All-cause mortality risk was analysed with competing-risk regression models. Vascular calcification was analysed in biopsies of the arteria epigastrica inferior in 111 patients. RESULTS Patients in the middle tertile of CAC density had the highest CAC score, CAC volume, age, CVD, PEW, FRS, hsCRP and IL-6. In competing risk analysis, the middle {subhazard ratio [sHR] 10.7 [95% confidence interval (CI) 2.0-57.3]} and high [sHR 8.9 (95% CI 1.5-51.8)] tertiles of CAC density associated with increased mortality, independent of CAC volume. The high tertile of CAC volume, independent of CAC density, associated with increased mortality [sHR 8.9 (95% CI 1.5-51.8)]. Arterial media calcification was prominent and associated with CAC volume and CAC density. CONCLUSIONS In CKD5, mortality increased linearly with higher CAC score and CAC volume whereas for CAC density an inverse J-shaped pattern was observed, with the crude mortality rate being highest for the middle tertile of CAC density. CAC volume and CAC density were associated with the extent of arterial media calcification.
Collapse
Affiliation(s)
- Hideyuki Mukai
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Lu Dai
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Zhimin Chen
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Bengt Lindholm
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Jonaz Ripsweden
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Torkel B Brismar
- Division of Medical Imaging and Technology, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Olof Heimbürger
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Peter Barany
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Abdul Rashid Qureshi
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| | - Magnus Söderberg
- Department of Pathology, Drug Safety and Metabolism, AstraZeneca, Mölndal, Sweden
| | - Magnus Bäck
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
| | - Peter Stenvinkel
- Division of Renal Medicine and Baxter Novum, Department of Clinical Science, Intervention and Technology, Karolinska Institutet, Campus Flemingsberg, Stockholm, Sweden
| |
Collapse
|
188
|
Li Y, Sun W, Saaoud F, Wang Y, Wang Q, Hodge J, Hui Y, Yin S, Lessner SM, Kong X, Fan D. MiR155 modulates vascular calcification by regulating Akt-FOXO3a signalling and apoptosis in vascular smooth muscle cells. J Cell Mol Med 2020; 25:535-548. [PMID: 33210462 PMCID: PMC7810936 DOI: 10.1111/jcmm.16107] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/13/2020] [Accepted: 11/01/2020] [Indexed: 12/17/2022] Open
Abstract
microRNA‐155 (miR155) is pro‐atherogenic; however, its role in vascular calcification is unknown. In this study, we aim to examine whether miR155 regulates vascular calcification and to understand the underlying mechanism. Quantitative real‐time PCR showed that miR155 is highly expressed in human calcific carotid tissue and positively correlated with the expression of osteogenic genes. Wound‐healing assay and TUNEL staining showed deletion of miR155 inhibited vascular smooth muscle cell (VSMC) migration and apoptosis. miR155 deficiency attenuated calcification of cultured mouse VSMCs and aortic rings induced by calcification medium, whereas miR155 overexpression promoted VSMC calcification. Compared with wild‐type mice, miR155−/− mice showed significant resistance to vitamin D3 induced vascular calcification. Protein analysis showed that miR155 deficiency alleviated the reduction of Rictor, increased phosphorylation of Akt at S473 and accelerated phosphorylation and degradation of FOXO3a in cultured VSMCs and in the aortas of vitamin D3‐treated mice. A PI3K inhibitor that suppresses Akt phosphorylation increased, whereas a pan‐caspase inhibitor that suppresses apoptosis reduced VSMC calcification; and both inhibitors diminished the protective effects of miR155 deficiency on VSMC calcification. In conclusion, miR155 deficiency attenuates vascular calcification by increasing Akt phosphorylation and FOXO3a degradation, and thus reducing VSMC apoptosis induced by calcification medium.
Collapse
Affiliation(s)
- Yong Li
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Wei Sun
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.,Department of Cardiology and Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Fatma Saaoud
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Yuzhen Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Quanyi Wang
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Johnie Hodge
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Yvonne Hui
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Sophia Yin
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Susan M Lessner
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - Xiangqing Kong
- Department of Cardiology and Department of Cardiothoracic Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Daping Fan
- Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| |
Collapse
|
189
|
Donato M, Ferri N, Lupo MG, Faggin E, Rattazzi M. Current Evidence and Future Perspectives on Pharmacological Treatment of Calcific Aortic Valve Stenosis. Int J Mol Sci 2020; 21:ijms21218263. [PMID: 33158204 PMCID: PMC7663524 DOI: 10.3390/ijms21218263] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023] Open
Abstract
Calcific aortic valve stenosis (CAVS), the most common heart valve disease, is characterized by the slow progressive fibro-calcific remodeling of the valve leaflets, leading to progressive obstruction to the blood flow. CAVS is an increasing health care burden and the development of an effective medical treatment is a major medical need. To date, no effective pharmacological therapies have proven to halt or delay its progression to the severe symptomatic stage and aortic valve replacement represents the only available option to improve clinical outcomes and to increase survival. In the present report, the current knowledge and latest advances in the medical management of patients with CAVS are summarized, placing emphasis on lipid-lowering agents, vasoactive drugs, and anti-calcific treatments. In addition, novel potential therapeutic targets recently identified and currently under investigation are reported.
Collapse
Affiliation(s)
- Maristella Donato
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Nicola Ferri
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Maria Giovanna Lupo
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, 35122 Padova, Italy; (M.D.); (N.F.); (M.G.L.)
| | - Elisabetta Faggin
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
| | - Marcello Rattazzi
- Department of Medicine—DIMED, University of Padova, 35122 Padova, Italy;
- Correspondence: ; Tel.: +39-0498-211-867 or +39-0422-322-207
| |
Collapse
|
190
|
Tianma Gouteng Decoction Exerts Cardiovascular Protection by Upregulating OPG and TRAIL in Spontaneously Hypertensive Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:3439191. [PMID: 33133215 PMCID: PMC7593748 DOI: 10.1155/2020/3439191] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 09/29/2020] [Accepted: 10/10/2020] [Indexed: 11/24/2022]
Abstract
Tianma Gouteng Decoction (TGD) is widely used in traditional Chinese medicine for the treatment of hypertension and its related complications, but its mechanisms remain incompletely defined. We now aim to assess the protective effect of TGD against cardiovascular damage and to investigate its characteristics and underlying mechanisms. Blood pressure was determined in TGD-treated spontaneously hypertensive rats (SHR) by noninvasive measurements. Echocardiography was performed to assess cardiac function and structure and sirius red staining to evaluate cardiac fibrosis, and the degree of vascular remodeling was evaluated. Additionally, vasoconstriction and relaxation factor expression changes were examined by means of ELISA. Protein expression changes were verified by western blot. Compared with untreated SHR, TGD-treated SHR exhibited cardiovascular traits more akin to those of the normotensive Wistar Kyoto (WKY) rats. That is, they had lower diastolic blood pressure, systolic blood pressure and mean BP, and increased expression of vasodilation factor. We also found that TGD reduces ventricular and vascular remodeling and improves cardiac function in SHR. Finally, we tested the antiapoptosis effect TGD exerts in SHR, ostensibly by upregulating the expression of OPG, TRAIL, and death receptor 5 (DR5) and downregulating caspases 8, 7, and 3. TRAIL may also exert antiapoptotic and prosurvival effects by upregulating AKT expression. Therefore, TGD may reverse cardiovascular remodeling in SHR by upregulating the expression of OPG and TRAIL, upregulating AKT, and inhibiting apoptosis, at least in part. For the first time, we have shown that OPG and TRAIL play complimentary cardioprotective roles in SHR.
Collapse
|
191
|
Lin WC, Tsai JP, Lai YH, Lin YL, Kuo CH, Wang CH, Hsu BG. Serum osteoprotegerin level is positively associated with peripheral artery disease in patients with peritoneal dialysis. Ren Fail 2020; 42:131-136. [PMID: 31950864 PMCID: PMC7006676 DOI: 10.1080/0886022x.2020.1714654] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 12/30/2019] [Accepted: 01/01/2020] [Indexed: 12/25/2022] Open
Abstract
Osteoprotegerin (OPG) is a potential biomarker of cardiovascular disease complications and severity. Peripheral arterial disease (PAD) is associated with an increased risk of death in patients on peritoneal dialysis (PD). Therefore, this study aimed to evaluate the relationship between serum OPG levels and PAD by measuring the ankle-brachial index (ABI) of patients on PD. A commercial enzyme-linked immunosorbent assay kit was used to measure OPG values. Left or right ABI values of <0.9 were categorized as the low ABI group. Among 70 patients on PD, 13 (18.6%) were categorized in the low ABI group. Patients in the low ABI group had higher prevalence of diabetes mellitus (p = .044) and higher serum C-reactive protein (CRP) (p < .001) and OPG levels (p < .001) but lower creatinine (p = .013) and peritoneal Kt/V (p = .048) levels than those in the normal ABI group. Results of multivariable logistic regression analysis revealed that OPG [adjusted odds ratio (aOR) 1.027, 95% confidence interval (CI) 1.010-1.045, p = .002] and CRP (aOR 1.102, 95% CI 1.006-1.207, p = .037) levels were independent predictors of PAD in patients on PD. OPG can also be used to predict PAD development with the area under the receiver operating characteristic curve of 0.823 (95% CI: 0.714-0.904, p < .001) in patients on PD. Therefore, serum OPG and CRP levels can be considered as risk factors for PAD development in patients on PD.
Collapse
Affiliation(s)
- Wei-Chen Lin
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Jen-Pi Tsai
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Division of Nephrology, Department of Internal Medicine, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Yu-Hsien Lai
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Yu-Li Lin
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chiu-Huang Kuo
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Chih-Hsien Wang
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Bang-Gee Hsu
- Division of Nephrology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| |
Collapse
|
192
|
Stompór T. An Overview of the Pathophysiology of Vascular Calcification in Chronic Kidney Disease. Perit Dial Int 2020. [DOI: 10.1177/089686080702702s37] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Abnormalities of calcium–phosphate balance, with subsequent bone metabolism disorders, are among the key and earliest features of chronic kidney disease (CKD). Recently, another consequence of these abnormalities was brought to light—namely, vascular calcification. Most studies performed in patients on dialysis suggest that their vascular calcification is more advanced than that seen in the general population. Furthermore, the progression of vessel wall mineralization is much more dynamic in patients with CKD. Apart from the commonly assessed factors that promote vascular calcification, such as age, duration of dialysis, or poor control of calcium–phosphate status, several other factors have recently been identified. In the spectrum of substances involved in the regulation of the process of soft-tissue calcification, the most extensively studied in the nephrology literature are bone morphogenetic protein 7, osteoprotegerin, matrix Gla protein, fetuin-A, and the phosphatonins. Better understanding of the mechanisms underlying excess vascular mineralization have led to the development of promising new therapies.
Collapse
Affiliation(s)
- Tomasz Stompór
- Chair and Department of Nephrology, Medical Faculty, Jagiellonian University, Cracow, Poland
| |
Collapse
|
193
|
Abstract
The phenotypic trait of high bone mass (HBM) is an excellent example of the nexus between common and rare disease genetics. HBM may arise from carriage of many 'high bone mineral density [BMD]'-associated alleles, and certainly the genetic architecture of individuals with HBM is enriched with high BMD variants identified through genome-wide association studies of BMD. HBM may also arise as a monogenic skeletal disorder, due to abnormalities in bone formation, bone resorption, and/or bone turnover. Individuals with monogenic disorders of HBM usually, though not invariably, have other skeletal abnormalities (such as mandible enlargement) and thus are best regarded as having a skeletal dysplasia rather than just isolated high BMD. A binary etiological division of HBM into polygenic vs. monogenic, however, would be excessively simplistic: the phenotype of individuals carrying rare variants of large effect can still be modified by their common variant polygenic background, and by the environment. HBM disorders-whether predominantly polygenic or monogenic in origin-are not only interesting clinically and genetically: they provide insights into bone processes that can be exploited therapeutically, with benefits both for individuals with these rare bone disorders and importantly for the many people affected by the commonest bone disease worldwide-i.e., osteoporosis. In this review we detail the genetic architecture of HBM; we provide a conceptual framework for considering HBM in the clinical context; and we discuss monogenic and polygenic causes of HBM with particular emphasis on anabolic causes of HBM.
Collapse
Affiliation(s)
- Celia L. Gregson
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Emma L. Duncan
- Department of Twin Research & Genetic Epidemiology, School of Life Course Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
194
|
Pertusa C, Tarín JJ, Cano A, García-Pérez MA. Association of a single nucleotide polymorphism of RANK gene with blood pressure in Spanish women. Medicine (Baltimore) 2020; 99:e22436. [PMID: 33019425 PMCID: PMC7535656 DOI: 10.1097/md.0000000000022436] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/31/2020] [Accepted: 08/23/2020] [Indexed: 12/30/2022] Open
Abstract
In addition to governing key functions in bone metabolism and the immune system, the RANK/RANKL/OPG system plays a role in the vascular system, particularly in vascular calcification and atherosclerosis.Given that these 2 phenotypes are considered a major cause of high blood pressure (BP), in this study we analyzed the association of SNPs in RANK and OPG genes with blood pressure. An observational study was conducted of 2 SNPs in the RANK gene (rs884205 and rs78326403) and 1 in the OPG gene (rs4876869) with systolic (SBP) and diastolic blood pressure (DBP) in a cohort of 695 women.Data analysis revealed a statistically significant association between the SNP rs884205 and BP pressure (SBP and DBP). Analyzing this relationship by the dominant inheritance model for this SNP (allele risk: A), women of the AA/AC genotype showed higher BP than women of the CC genotype, both for SBP (P = .001) and for DBP (P = .003), and these associations both surpassed the Bonferroni threshold for multiple comparisons. Multivariate regression analysis including known predictors of BP as independent variables was performed to evaluate the strength of this association, which in the case of the SNP rs884205 of the RANK gene remained statistically significant after adjustment for both SBP (P = .0006) and DBP (P = .005), demonstrating the key role of this SNP in BP.We report a robust association between the SNP rs884205 in RANK gene and BP in women, and this SNP is validated as a candidate in cardiovascular risk studies.
Collapse
Affiliation(s)
- Clara Pertusa
- Research Foundation, INCLIVA Institute of Health Research, Valencia
| | - Juan J. Tarín
- Department of Cellular Biology, Functional Biology and Physical Anthropology, University of Valencia, Burjassot
| | - Antonio Cano
- Department of Pediatrics, Obstetrics and Gynecology, University of Valencia, Valencia
| | - Miguel Angel García-Pérez
- Research Foundation, INCLIVA Institute of Health Research, Valencia
- Department of Genetics, University of Valencia, Burjassot, Spain
| |
Collapse
|
195
|
Uremic serum-induced calcification of human aortic smooth muscle cells is a regulated process involving Klotho and RUNX2. Biosci Rep 2020; 39:BSR20190599. [PMID: 31519772 PMCID: PMC6822531 DOI: 10.1042/bsr20190599] [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] [Subscribe] [Scholar Register] [Received: 02/24/2019] [Revised: 07/16/2019] [Accepted: 07/24/2019] [Indexed: 01/17/2023] Open
Abstract
Vascular calcification (VC) is common in subjects with chronic kidney disease (CKD) and is associated with increased cardiovascular risk. It is an active process involving transdifferentiation of arterial smooth muscle cells (SMCs) into osteogenic phenotype. We investigated the ability of serum from CKD subjects to induce calcification in human SMCs in vitro (calcific potential of sera: CP), and associated changes in expression of Runt-related transcription factor 2 (RUNX2), SM22α, and Klotho. Sera from subjects with CKD (18 stage 3, 17 stage 4/5, and 29 stage 5D) and 20 controls were added to human cultured SMCs and CP quantified. The CP of CKD sera was greater (P<0.01) than that of controls, though not influenced by CKD stage. Modification of diet in renal disease estimated glomerular filtration rate (MDRD-4 eGFR) (P<0.001), serum phosphate (P=0.042), receptor activator of nuclear factor κappa-B ligand (RANKL) (P=0.001), parathyroid hormone (PTH) (P=0.014), and high-density lipoprotein (HDL)/cholesterol ratio (P=0.026) were independent predictors of CP accounting for 45% of variation. Adding calcification buffer (CB: calcium chloride [7 mM] and β-glycerophosphate [7 mM]) increased the CP of control sera to approximate that of CKD sera. CP of CKD sera was unchanged. CKD sera increased RUNX2 expression (P<0.01) in human SMCs and decreased SM22α expression (P<0.05). Co-incubating control but not CKD serum with CB further increased RUNX2 expression (P<0.01). Both SM22α and Klotho expression decreased significantly (P<0.01) in the presence of CKD serum, and were virtually abolished with stage 5D sera. These findings support active regulation by CKD serum of in vitro VC by induction of RUNX2 and suppression of SM22α and Klotho.
Collapse
|
196
|
Ferreira GZ, Zen Filho EV, Rubira-Bullen IRF, Garlet GP, Santos CF, Santos PSDS. Delayed alveolar bone repair and osteonecrosis associated with Zoledronic Acid therapy in rats: macroscopic, microscopic and molecular analysis. J Appl Oral Sci 2020; 28:e20200204. [PMID: 32997086 PMCID: PMC7518245 DOI: 10.1590/1678-7757-2020-0204] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2020] [Accepted: 07/14/2020] [Indexed: 12/16/2022] Open
Abstract
Objective This study aims to evaluate bone repair and the development of the medication related osteonecrosis of the jaw (MRONJ) associated with the use of zoledronic acid in Wistar rats. Methodology 48 male Wistar rats were divided into four groups: ZA, treated with intraperitoneal zoledronic acid, 0.6 mg/kg every 28 days, totaling five doses; control (C), treated with 0.9% sodium chloride; ZA-surgical (SZA) and C-surgical (SC), submitted to extraction of the right upper molars 45 days after the first application. Alveolar bone repair was evaluated by macroscopic and histological analysis. Protein expression evaluations were performed by qPCR. Results Macroscopic evaluation showed that 91.66% (11) of the animals in the SZA group and 41.66% (5) from the SC group presented solution of epithelium continuity (P<0.05). All animals in the SZA group and none in the SC group had bone sequestration. The area of osteonecrosis was higher in the SZA group than in the SC group (P<0.05). In molecular evaluation, the SZA group presented changes in the expression of markers for osteoclasts, with increased RANK and RANKL, and a decrease in OPG. Conclusion The results highlighted strong and evident interference of zoledronic acid in bone repair of the socket, causing osteonecrosis and delayed bone remodeling.
Collapse
Affiliation(s)
- Gustavo Zanna Ferreira
- UniCesumar - Departamento de Odontologia, Maringá, PR, Brasil; Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Cirurgia, Estomatologia, Patologia e Radiologia, Bauru, SP, Brasil
| | | | - Izabel Regina Fisher Rubira-Bullen
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Cirurgia, Estomatologia, Patologia e Radiologia, Bauru, SP, Brasil
| | - Gustavo Pompermaier Garlet
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Carlos Ferreira Santos
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Ciências Biológicas, Bauru, SP, Brasil
| | - Paulo Sérgio da Silva Santos
- Universidade de São Paulo, Faculdade de Odontologia de Bauru, Departamento de Cirurgia, Estomatologia, Patologia e Radiologia, Bauru, SP, Brasil
| |
Collapse
|
197
|
Sarker H, Hardy E, Haimour A, Karim MA, Scholl-Bürgi S, Martignetti JA, Botto LD, Fernandez-Patron C. Comparative Serum Analyses Identify Cytokines and Hormones Commonly Dysregulated as Well as Implicated in Promoting Osteolysis in MMP-2-Deficient Mice and Children. Front Physiol 2020; 11:568718. [PMID: 33101055 PMCID: PMC7546215 DOI: 10.3389/fphys.2020.568718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 09/08/2020] [Indexed: 11/13/2022] Open
Abstract
Deficiency of matrix metalloproteinase 2 (MMP-2) causes a complex syndrome characterized by multicentric osteolysis, nodulosis, and arthropathy (MONA) as well as cardiac valve defects, dwarfism and hirsutism. MMP-2 deficient (Mmp2 -/-) mice are a model for this rare multisystem pediatric syndrome but their phenotype remains incompletely characterized. Here, we extend the phenotypic characterization of MMP-2 deficiency by comparing the levels of cytokines and chemokines, soluble cytokine receptors, angiogenesis factors, bone development factors, apolipoproteins and hormones in mice and humans. Initial screening was performed on an 8-year-old male presenting a previously unreported deletion mutation c1294delC (Arg432fs) in the MMP2 gene and diagnosed with MONA. Of eighty-one serum biomolecules analyzed, eleven were upregulated (>4-fold), two were downregulated (>4-fold) and sixty-eight remained unchanged, compared to unaffected controls. Specifically, Eotaxin, GM-CSF, M-CSF, GRO-α, MDC, IL-1β, IL-7, IL-12p40, MIP-1α, MIP-1β, and MIG were upregulated and epidermal growth factor (EGF) and ACTH were downregulated in this patient. Subsequent analysis of five additional MMP-2 deficient patients confirmed the upregulation in Eotaxin, IL-7, IL-12p40, and MIP-1α, and the downregulation in EGF. To establish whether these alterations are bona fide phenotypic traits of MMP-2 deficiency, we further studied Mmp2 -/- mice. Among 32 cytokines measured in plasma of Mmp2 -/- mice, the cytokines Eotaxin, IL-1β, MIP-1α, and MIG were commonly upregulated in mice as well as patients with MMP-2 deficiency. Moreover, bioactive cortisol (a factor that exacerbates osteoporosis) was also elevated in MMP-2 deficient mice and patients. Among the factors we have identified to be dysregulated in MMP-2 deficiency many are osteoclastogenic and could potentially contribute to bone disorder in MONA. These new molecular phenotypic traits merit being targeted in future research aimed at understanding the pathological mechanisms elicited by MMP-2 deficiency in children.
Collapse
Affiliation(s)
- Hassan Sarker
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | | | - Ayman Haimour
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Mahmoud A. Karim
- Department of Cell Biology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Sabine Scholl-Bürgi
- Clinic for Pediatrics I, Inherited Metabolic Disorders, Medical University of Innsbruck, Innsbruck, Austria
| | - John A. Martignetti
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
- Rudy L. Ruggles Biomedical Research Institute, Nuvance Health, Danbury, CT, United States
| | - Lorenzo D. Botto
- Department of Pediatrics, Division of Medical Genetics and Pediatrics, The University of Utah, Salt Lake City, UT, United States
| | - Carlos Fernandez-Patron
- Department of Biochemistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
198
|
Abstract
During menopausal transition, decreased level of estrogen brings a number of physiological problems and hormonal changes. In this study, promoter methylation of RANKL and FSHR genes were identified in 30 premenopausal and 35 postmenopausal women using methylation-specific high resolution melting (MS-HRM) analysis. The statistical analyses and their association with patient characteristics were performed by Pearson χ2 and Fisher's exact test (p <0.05). The methylated RANKL gene was detected in 16 postmenopausal cases, and 12 (75.0%) of the RANKL methylated cases had hot flashes (p = 0.024). The methylated FSHR gene was detected in 18 postmenopausal cases, and 13 (75.0%) of the methylated cases had hot flashes (p = 0.028). In vitro studies demonstrated the association between RANKL expression, FSH level and hot flashes in the mouse. Although lack of epigenetic studies in this field proves our results crucial and therefore, our results showed magnitude of epigenetic profiles of Turkish Cypriot post-menopausal women. This was the first study which has investigated the RANKL and FSHR methylation and their relationship with hot flashes in postmenopausal women.
Collapse
|
199
|
The top 100 highly cited articles on osteoporosis from 1990 to 2019: a bibliometric and visualized analysis. Arch Osteoporos 2020; 15:144. [PMID: 32935223 DOI: 10.1007/s11657-020-0705-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Accepted: 01/27/2020] [Indexed: 02/05/2023]
Abstract
UNLABELLED Research on osteoporosis is a well-developed and promising research field. The top 100 literature included 73 articles and 27 reviews. The average citation number was 747 (range 370 to 2970). Researchers and institutions from the USA, the UK, and France contributed the most to the top high-cited articles. PURPOSE To provide a bibliometric and visualized analysis of the top 100 highly cited articles on osteoporosis indexed by the Web of Science (WoS) from 1990 to 2019. METHODS Data were obtained from the WoS Core Collection on Jan 10, 2020. Qualitative and quantitative analysis was conducted based on WoS. Collaboration analysis and keywords analysis were performed using VosView software. RESULTS A total of 12,863 references were obtained. The top 100 highly cited literature included 73 articles and 27 reviews. The average citation number of the 100 articles was 747 (range 370 to 2970). The fund sources mostly came from the USA. A total of 29 journals published the top 100 highly cited literature. The New England Journal of Medicine had the largest number of papers and the highest total cited times. The USA published 72 articles. The University of California San Francisco published 17 articles, followed by University of Sheffield and Mayo Clinic and Mayo Foundation. Cooper C had the most publications (n = 12) and Genant HK had the largest total citation (n = 11,055). Considering only the first author and corresponding author, Kanis JA had published the most articles. Researchers and institutions from the USA, the UK, and France contributed the most to the top high-cited articles. CONCLUSIONS Research on osteoporosis is a well-developed and promising research field. The top 100 articles have been cited widely and actively. New England Journal of Medicine was the most popular journal. The most productive country was the USA. The University of California San Francisco, University of Sheffield, and Mayo Clinic and Mayo Foundation were the most productive institutions. Cooper C, Kanis JA, and Genant HK were the most prolific and influential authors. Researchers and institutions from North America and Europe contributed the most.
Collapse
|
200
|
Kim JM, Lin C, Stavre Z, Greenblatt MB, Shim JH. Osteoblast-Osteoclast Communication and Bone Homeostasis. Cells 2020; 9:E2073. [PMID: 32927921 PMCID: PMC7564526 DOI: 10.3390/cells9092073] [Citation(s) in RCA: 638] [Impact Index Per Article: 127.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/08/2020] [Accepted: 09/08/2020] [Indexed: 12/23/2022] Open
Abstract
Bone remodeling is tightly regulated by a cross-talk between bone-forming osteoblasts and bone-resorbing osteoclasts. Osteoblasts and osteoclasts communicate with each other to regulate cellular behavior, survival and differentiation through direct cell-to-cell contact or through secretory proteins. A direct interaction between osteoblasts and osteoclasts allows bidirectional transduction of activation signals through EFNB2-EPHB4, FASL-FAS or SEMA3A-NRP1, regulating differentiation and survival of osteoblasts or osteoclasts. Alternatively, osteoblasts produce a range of different secretory molecules, including M-CSF, RANKL/OPG, WNT5A, and WNT16, that promote or suppress osteoclast differentiation and development. Osteoclasts also influence osteoblast formation and differentiation through secretion of soluble factors, including S1P, SEMA4D, CTHRC1 and C3. Here we review the current knowledge regarding membrane bound- and soluble factors governing cross-talk between osteoblasts and osteoclasts.
Collapse
Affiliation(s)
- Jung-Min Kim
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (J.-M.K.); (C.L.); (Z.S.)
| | - Chujiao Lin
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (J.-M.K.); (C.L.); (Z.S.)
| | - Zheni Stavre
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (J.-M.K.); (C.L.); (Z.S.)
| | - Matthew B. Greenblatt
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY 10065, USA;
| | - Jae-Hyuck Shim
- Division of Rheumatology, Department of Medicine, University of Massachusetts Medical School, Worcester, MA 01605, USA; (J.-M.K.); (C.L.); (Z.S.)
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|