1
|
Chandra A, Law SF, Pignolo RJ. Changing landscape of hematopoietic and mesenchymal cells and their interactions during aging and in age-related skeletal pathologies. Mech Ageing Dev 2025; 225:112059. [PMID: 40220914 PMCID: PMC12103995 DOI: 10.1016/j.mad.2025.112059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 03/26/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
Aging profoundly impacts mesenchymal and hematopoietic lineage cells, including their progenitors-the skeletal stem cells (SSCs) and hematopoietic stem cells (HSCs), respectively. SSCs are crucial for skeletal development, homeostasis, and regeneration, maintaining bone integrity by differentiating into osteoblasts, adipocytes, and other lineages that contribute to the bone marrow (BM) microenvironment. Meanwhile, HSCs sustain hematopoiesis and immune function. With aging, SSCs and HSCs undergo significant functional decline, partly driven by cellular senescence-a hallmark of aging characterized by irreversible growth arrest, secretion of pro-inflammatory factors (senescence associated secretory phenotype, SASP), and impaired regenerative potential. In SSCs, senescence skews lineage commitment toward adipogenesis at the expense of osteogenesis, contributing to increased bone marrow adiposity , reduced bone quality, and osteoporosis. Similarly, aged HSCs exhibit diminished self-renewal, biased differentiation, and heightened inflammation, compromising hematopoietic output and immune function. In this review, we examine the age-related cellular and molecular changes in SSCs and HSCs, their lineage decisions in the aging microenvironment, and the interplay between skeletal and hematopoietic compartments. We also discuss the role of senescence-driven alterations in BM homeostasis and how targeting cellular aging mechanisms may offer therapeutic strategies for mitigating age-related skeletal and hematopoietic decline.
Collapse
Affiliation(s)
- Abhishek Chandra
- Department of Physiology and Biomedical Engineering, USA; Department of Medicine, Divisions of Hospital Internal Medicine and Section on Geriatric Medicine and Gerontology, USA; Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, MN, USA.
| | - Susan F Law
- Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, MN, USA
| | - Robert J Pignolo
- Department of Physiology and Biomedical Engineering, USA; Department of Medicine, Divisions of Hospital Internal Medicine and Section on Geriatric Medicine and Gerontology, USA; Robert and Arlene Kogod Aging Center, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
2
|
Weaver SR, Torres HM, Arnold KM, Zars EL, Peralta-Herrera E, Taylor EL, Yu K, Marron Fernandez de Velasco E, Wickman K, McGee-Lawrence ME, Bradley EW, Westendorf JJ. Girk3 deletion increases osteoblast maturation and bone mass accrual in adult male mice. JBMR Plus 2024; 8:ziae108. [PMID: 39228688 PMCID: PMC11370632 DOI: 10.1093/jbmrpl/ziae108] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 07/25/2024] [Indexed: 09/05/2024] Open
Abstract
Osteoporosis and other metabolic bone diseases are prevalent in the aging population. While bone has the capacity to regenerate throughout life, bone formation rates decline with age and contribute to reduced bone density and strength. Identifying mechanisms and pathways that increase bone accrual in adults could prevent fractures and accelerate healing. G protein-gated inwardly rectifying K+ (GIRK) channels are key effectors of G protein-coupled receptor signaling. Girk3 was recently shown to regulate endochondral ossification. Here, we demonstrate that deletion of Girk3 increases bone mass after 18 weeks of age. Male 24-week-old Girk3 -/- mice have greater trabecular bone mineral density and bone volume fraction than wildtype (WT) mice. Osteoblast activity is moderately increased in 24-week-old Girk3 -/- mice compared to WT mice. In vitro, Girk3-/- bone marrow stromal cells (BMSCs) are more proliferative than WT BMSCs. Calvarial osteoblasts and BMSCs from Girk3 -/- mice are also more osteogenic than WT cells, with altered expression of genes that regulate the wingless-related integration site (Wnt) family. Wnt inhibition via Dickkopf-1 (Dkk1) or β-catenin inhibition via XAV939 prevents enhanced mineralization, but not proliferation, in Girk3 -/- BMSCs and slows these processes in WT cells. Finally, selective ablation of Girk3 from cells expressing Cre recombinase from the 2.3 kb-Col1a1 promoter, including osteoblasts and osteocytes, is sufficient to increase bone mass and bone strength in male mice at 24 weeks of age. Taken together, these data demonstrate that Girk3 regulates progenitor cell proliferation, osteoblast differentiation, and bone mass accrual in adult male mice.
Collapse
Affiliation(s)
- Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | - Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | - Elizabeth L Zars
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | | | | | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN 55455, United States
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University, Augusta, GA 30912, United States
| | - Elizabeth W Bradley
- Department of Orthopedics, School of Medicine, University of Minnesota, Minneapolis, MN 55455, United States
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
3
|
Yeo D, Zars Fisher EL, Khosla S, Farr JN, Westendorf JJ. Hdac3-deficiency increases senescence-associated distention of satellite DNA and telomere-associated foci in osteoprogenitor cells. J Bone Miner Res 2024; 39:994-1007. [PMID: 38843356 PMCID: PMC12102593 DOI: 10.1093/jbmr/zjae085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 05/04/2024] [Accepted: 06/05/2024] [Indexed: 08/07/2024]
Abstract
Histone deacetylase 3 (Hdac3) is an epigenetic regulator of gene expression and interacts with skeletal transcription factors such as Runx2. We previously reported that conditional deletion of Hdac3 in Osterix-Cre recombinase-expressing osteoprogenitor cells (Hdac3 CKOOsx) caused osteopenia and increased marrow adiposity, both hallmarks of skeletal aging. We also showed that Runx2+ cells within osteogenic cultures of Hdac3-depleted bone marrow stromal cells (BMSCs) contain lipid droplets (LDs). Cellular senescence, a nonproliferative metabolically active state, is associated with increased marrow adiposity, bone loss, and aging. In this study, we sought to determine if Hdac3 depleted Runx2+ pre-osteoblasts from young mice exhibit chromatin changes associated with early cellular senescence and how these events correlate with the appearance of LDs. We first confirmed that BMSCs from Hdac3 CKOOsx mice have more Runx2 + LD+ cells compared with controls under osteogenic conditions. We then measured senescence-associated distention of satellite (SADS) DNA and telomere-associated foci (TAFs) in Hdac3 CKOOsx and control BMSCs. In situ, Runx2+ cells contained more SADS per nuclei in Hdac3 CKOOsx femora than in controls. Runx2+ BMSCs from Hdac3 CKOOsx mice also contained more SADS and TAFs per nuclei than Runx2+ cells from age-matched control mice in vitro. SADs and TAFs were present at similar levels in Runx2 + LD+ cells and Runx2 + LD- cells from Hdac3 CKOOsx mice. Hdac inhibitors also increased the number of SADS in Runx2 + LD+ and Runx2 + LD- WT BMSCs. Senolytics reduced viable cell numbers in Hdac3 CKOOsx BMSC cultures. These data demonstrate that the depletion of Hdac3 in osteochondral progenitor cells triggers LD formation and early events in cellular senescence in Runx2+ BMSCs through mutually exclusive mechanisms.
Collapse
Affiliation(s)
- Dongwook Yeo
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
| | | | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States
| | - Joshua N Farr
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, United States
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, United States
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, United States
| |
Collapse
|
4
|
Raimondi L, De Luca A, Gallo A, Perna F, Cuscino N, Cordaro A, Costa V, Bellavia D, Faldini C, Scilabra SD, Giavaresi G, Toscano A. Investigating the Differential Circulating microRNA Expression in Adolescent Females with Severe Idiopathic Scoliosis: A Proof-of-Concept Observational Clinical Study. Int J Mol Sci 2024; 25:570. [PMID: 38203740 PMCID: PMC10779108 DOI: 10.3390/ijms25010570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/28/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Adolescent Idiopathic Scoliosis (AIS) is the most common form of three-dimensional spinal disorder in adolescents between the ages of 10 and 18 years of age, most commonly diagnosed in young women when severe disease occurs. Patients with AIS are characterized by abnormal skeletal growth and reduced bone mineral density. The etiology of AIS is thought to be multifactorial, involving both environmental and genetic factors, but to date, it is still unknown. Therefore, it is crucial to further investigate the molecular pathogenesis of AIS and to identify biomarkers useful for predicting curve progression. In this perspective, the relative abundance of a panel of microRNAs (miRNAs) was analyzed in the plasma of 20 AIS patients and 10 healthy controls (HC). The data revealed a significant group of circulating miRNAs dysregulated in AIS patients compared to HC. Further bioinformatic analyses evidenced a more restricted expression of some miRNAs exclusively in severe AIS females. These include some members of the miR-30 family, which are considered promising regulators for treating bone diseases. We demonstrated circulating extracellular vesicles (EVs) from severe AIS females contained miR-30 family members and decreased the osteogenic differentiation of mesenchymal stem cells. Proteomic analysis of EVs highlighted the expression of proteins associated with orthopedic disease. This study provides preliminary evidence of a miRNAs signature potentially associated with severe female AIS and suggests the corresponding vesicular component may affect cellular mechanisms crucial in AIS, opening the scenario for in-depth studies on prognostic differences related to gender and grade.
Collapse
Affiliation(s)
- Lavinia Raimondi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Angela De Luca
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Alessia Gallo
- Dipartimento di Ricerca, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Fabrizio Perna
- Ortopedia Generale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy (A.T.)
| | - Nicola Cuscino
- Dipartimento di Ricerca, IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), 90127 Palermo, Italy
| | - Aurora Cordaro
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Viviana Costa
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Daniele Bellavia
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Cesare Faldini
- Clinica Ortopedica e Traumatologica I, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy
| | - Simone Dario Scilabra
- Fondazione Ri.MED, Dipartimento di Ricerca IRCCS ISMETT, Via Ernesto Tricomi 5, 90145 Palermo, Italy
| | - Gianluca Giavaresi
- Scienze e Tecnologie Chirurgiche, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy; (L.R.)
| | - Angelo Toscano
- Ortopedia Generale, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano, 1/10, 40136 Bologna, Italy (A.T.)
| |
Collapse
|
5
|
Karkache IY, Molstad DHH, Vu E, Jensen ED, Bradley EW. Phlpp1 Expression in Osteoblasts Plays a Modest Role in Bone Homeostasis. JBMR Plus 2023; 7:e10806. [PMID: 38130760 PMCID: PMC10731110 DOI: 10.1002/jbm4.10806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 12/23/2023] Open
Abstract
Prior work demonstrated that Phlpp1 deficiency alters limb length and bone mass, but the cell types involved and requirement of Phlpp1 for this effect were unclear. To understand the function of Phlpp1 within bone-forming osteoblasts, we crossed Phlpp1 floxed mice with mice harboring type 1 collagen (Col1a12.3kb)-Cre. Mineralization of bone marrow stromal cell cultures derived from Phlpp1 cKOCol1a1 was unchanged, but levels of inflammatory genes (eg, Ifng, Il6, Ccl8) and receptor activator of NF-κB ligand/osteoprotegerin (RANKL/OPG) ratios were enhanced by either Phlpp1 ablation or chemical inhibition. Micro-computed tomography of the distal femur and L5 vertebral body of 12-week-old mice revealed no alteration in bone volume per total volume, but compromised femoral bone microarchitecture within Phlpp1 cKOCol1a1 conditional knockout females. Bone histomorphometry of the proximal tibia documented no changes in osteoblast or osteoclast number per bone surface but slight reductions in osteoclast surface per bone surface. Overall, our data show that deletion of Phlpp1 in type 1 collagen-expressing cells does not significantly alter attainment of peak bone mass of either males or females, but may enhance inflammatory gene expression and the ratio of RANKL/OPG. Future studies examining the role of Phlpp1 within models of advanced age, inflammation, or osteocytes, as well as functional redundancy with the related Phlpp2 isoform are warranted. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Ismael Y Karkache
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
- College of Veterinary SciencesUniversity of MinnesotaMinneapolisMNUSA
| | - David HH Molstad
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
| | - Elizabeth Vu
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
| | | | - Elizabeth W Bradley
- Department of OrthopedicsUniversity of MinnesotaMinneapolisMNUSA
- College of Veterinary SciencesUniversity of MinnesotaMinneapolisMNUSA
- Department of Orthopedic SurgeryStem Cell Institute, University of MinnesotaMinneapolisMNUSA
| |
Collapse
|
6
|
Dashti P, Thaler R, Hawse JR, Galvan ML, van der Eerden BJ, van Wijnen AJ, Dudakovic A. G-protein coupled receptor 5C (GPRC5C) is required for osteoblast differentiation and responds to EZH2 inhibition and multiple osteogenic signals. Bone 2023; 176:116866. [PMID: 37558192 PMCID: PMC10962865 DOI: 10.1016/j.bone.2023.116866] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/24/2023] [Accepted: 08/03/2023] [Indexed: 08/11/2023]
Abstract
Osteoblast differentiation is epigenetically suppressed by the H3K27 methyltransferase EZH2, and induced by the morphogen BMP2 and transcription factor RUNX2. These factors also regulate distinct G protein coupled receptors (GPRCs; e.g., PTH1R, GPR30/GPER1). Because GPRCs transduce many physiological stimuli, we examined whether BMP2 or EZH2 inhibition (i.e., GSK126) regulates other GPRC genes in osteoblasts. RNA-seq screening of >400 mouse GPRC-related genes showed that many GPRCs are downregulated during osteogenic differentiation. The orphan receptor GPRC5C, along with a small subset of other GPRCs, is induced by BMP2 or GSK126 during Vitamin C dependent osteoblast differentiation, but not by all-trans retinoic acid. ChIP-seq analysis revealed that GSK126 reduces H3K27me3 levels at the GPRC5C gene locus in differentiating MC3T3-E1 osteoblasts, consistent with enhanced GPRC5C mRNA expression. Loss of function analyses revealed that shRNA-mediated depletion of GPRC5C decreases expression of bone markers (e.g., BGLAP and IBSP) and mineral deposition in response to BMP2 or GSK126. GPRC5C mRNA was found to be reduced in the osteopenic bones of KLF10 null mice which have compromised BMP2 signaling. GPRC5C mRNA is induced by the bone-anabolic activity of 17β-estradiol in trabecular but not cortical bone following ovariectomy. Collectively, these findings suggest that GPRC5C protein is a key node in a pro-osteogenic axis that is normally suppressed by EZH2-mediated H3K27me3 marks and induced during osteoblast differentiation by GSK126, BMP2, and/or 17β-estradiol. Because GPRC5C protein is an understudied orphan receptor required for osteoblast differentiation, identification of ligands that induce GPRC5C signaling may support therapeutic strategies to mitigate bone-related disorders.
Collapse
Affiliation(s)
- Parisa Dashti
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - John R Hawse
- Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - M Lizeth Galvan
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Bram J van der Eerden
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Andre J van Wijnen
- Department of Internal Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands; Department of Biochemistry, University of Vermont, Burlington, VT, USA.
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA; Department of Biochemistry & Molecular Biology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
7
|
Taylor EL, Weaver SR, Lorang IM, Arnold KM, Bradley EW, Marron Fernandez de Velasco E, Wickman K, Westendorf JJ. GIRK3 deletion facilitates kappa opioid signaling in chondrocytes, delays vascularization and promotes bone lengthening in mice. Bone 2022; 159:116391. [PMID: 35314385 PMCID: PMC9035100 DOI: 10.1016/j.bone.2022.116391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/14/2022] [Indexed: 11/29/2022]
Abstract
Long bones are formed and repaired through the process of endochondral ossification. Activation of G protein-coupled receptor (GPCR) signaling pathways is crucial for skeletal development and long bone growth. G protein-gated inwardly-rectifying K+ (GIRK) channel genes are key functional components and effectors of GPCR signaling pathways in excitable cells of the heart and brain, but their roles in non-excitable cells that directly contribute to endochondral bone formation have not been studied. In this study, we analyzed skeletal phenotypes of Girk2-/-, Girk3-/- and Girk2/3-/- mice. Bones from 12-week-old Girk2-/- mice were normal in length, but femurs and tibiae from Girk3-/- and Girk2/3-/- mice were longer than age-matched controls at 12-weeks-old. Epiphyseal chondrocytes from 5-day-old Girk3-/- mice expressed higher levels of genes involved in collagen chain trimerization and collagen fibril assembly, lower levels of genes encoding VEGF receptors, and produced larger micromasses than wildtype chondrocytes in vitro. Girk3-/- chondrocytes were also more responsive to the kappa opioid receptor (KOR) ligand dynorphin, as evidenced by greater pCREB expression, greater cAMP and GAG production, and upregulation of Col2a1 and Sox9 transcripts. Imaging studies showed that Kdr (Vegfr2) and endomucin expression was dramatically reduced in bones from young Girk3-/- mice, supporting a role for delayed vasculogenesis and extended postnatal endochondral bone growth. Together these data indicate that GIRK3 controls several processes involved in bone lengthening.
Collapse
Affiliation(s)
- Earnest L Taylor
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America; Department of Cell Biology, University of North Carolina, NC, United States of America
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America
| | - Ian M Lorang
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America; University of Washington School of Medicine, Seattle, WA, United States of America
| | - Katherine M Arnold
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, United States of America
| | - Elizabeth W Bradley
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America; Department of Orthopedic Surgery, University of Minnesota, Minneapolis, MN, United States of America
| | | | - Kevin Wickman
- Department of Pharmacology, University of Minnesota, Minneapolis, MN, United States of America
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, United States of America; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
8
|
Muñoz A, Docaj A, Ugarteburu M, Carriero A. Poor bone matrix quality: What can be done about it? Curr Osteoporos Rep 2021; 19:510-531. [PMID: 34414561 DOI: 10.1007/s11914-021-00696-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/27/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF THE REVIEW Bone's ability to withstand load resisting fracture and adapting to it highly depends on the quality of its matrix and its regulators. This review focuses on the contribution of bone quality to fracture resistance and possible therapeutic targets for skeletal fragility in aging and disease. RECENT FINDINGS The highly organized, hierarchical composite structure of bone extracellular matrix together with its (re)modeling mechanisms and microdamage dynamics determines its stiffness, strength, and toughness. Aging and disease affect the biological processes regulating bone quality, thus resulting in defective extracellular matrix and bone fragility. Targeted therapies are being developed to restore bone's mechanical integrity. However, their current limitations include low tissue selectivity and adverse side effects. Biological and mechanical insights into the mechanisms controlling bone quality, together with advances in drug delivery and studies in animal models, will accelerate the development and translation to clinical application of effective targeted-therapeutics for bone fragility.
Collapse
Affiliation(s)
- Asier Muñoz
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA
| | - Anxhela Docaj
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA
| | - Maialen Ugarteburu
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA
| | - Alessandra Carriero
- Department of Biomedical Engineering, The City College of New York, 160 Convent Avenue, Steinman Bldg. Room 403C, New York, NY, 10031, USA.
| |
Collapse
|
9
|
Torres HM, VanCleave AM, Vollmer M, Callahan DL, Smithback A, Conn JM, Rodezno-Antunes T, Gao Z, Cao Y, Afeworki Y, Tao J. Selective Targeting of Class I Histone Deacetylases in a Model of Human Osteosarcoma. Cancers (Basel) 2021; 13:4199. [PMID: 34439353 PMCID: PMC8394112 DOI: 10.3390/cancers13164199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 02/01/2023] Open
Abstract
Dysregulation of histone deacetylases (HDACs) is associated with the pathogenesis of human osteosarcoma, which may present an epigenetic vulnerability as well as a therapeutic target. Domatinostat (4SC-202) is a next-generation class I HDAC inhibitor that is currently being used in clinical research for certain cancers, but its impact on human osteosarcoma has yet to be explored. In this study, we report that 4SC-202 inhibits osteosarcoma cell growth in vitro and in vivo. By analyzing cell function in vitro, we show that the anti-tumor effect of 4SC-202 involves the combined induction of cell-cycle arrest at the G2/M phase and apoptotic program, as well as a reduction in cell invasion and migration capabilities. We also found that 4SC-202 has little capacity to promote osteogenic differentiation. Remarkably, 4SC-202 revised the global transcriptome and induced distinct signatures of gene expression in vitro. Moreover, 4SC-202 decreased tumor growth of established human tumor xenografts in immunodeficient mice in vivo. We further reveal key targets regulated by 4SC-202 that contribute to tumor cell growth and survival, and canonical signaling pathways associated with progression and metastasis of osteosarcoma. Our study suggests that 4SC-202 may be exploited as a valuable drug to promote more effective treatment of patients with osteosarcoma and provide molecular insights into the mechanism of action of class I HDAC inhibitors.
Collapse
Affiliation(s)
- Haydee M. Torres
- Cancer Biology & Immunotherapies Group at Sanford Research, Sioux Falls, SD 57104, USA; (H.M.T.); (A.M.V.); (T.R.-A.); (Y.C.)
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA
| | - Ashley M. VanCleave
- Cancer Biology & Immunotherapies Group at Sanford Research, Sioux Falls, SD 57104, USA; (H.M.T.); (A.M.V.); (T.R.-A.); (Y.C.)
| | - Mykayla Vollmer
- Medical Student Research Program, University of South Dakota, Vermillion, SD 57069, USA;
| | - Dakota L. Callahan
- Sanford Program for Undergraduate Research, University of Sioux Falls, Sioux Falls, SD 57104, USA;
| | - Austyn Smithback
- Sanford PROMISE Scholar Program, Harrisburg High School, Sioux Falls, SD 57104, USA;
| | - Josephine M. Conn
- Sanford Program for Undergraduate Research, Carleton College, Northfield, MN 55057, USA;
| | - Tania Rodezno-Antunes
- Cancer Biology & Immunotherapies Group at Sanford Research, Sioux Falls, SD 57104, USA; (H.M.T.); (A.M.V.); (T.R.-A.); (Y.C.)
| | - Zili Gao
- Flow Cytometry Core at Sanford Research, Sioux Falls, SD 57104, USA;
| | - Yuxia Cao
- Cancer Biology & Immunotherapies Group at Sanford Research, Sioux Falls, SD 57104, USA; (H.M.T.); (A.M.V.); (T.R.-A.); (Y.C.)
| | - Yohannes Afeworki
- Functional Genomics & Bioinformatics Core Facility at Sanford Research, Sioux Falls, SD 57104, USA;
| | - Jianning Tao
- Cancer Biology & Immunotherapies Group at Sanford Research, Sioux Falls, SD 57104, USA; (H.M.T.); (A.M.V.); (T.R.-A.); (Y.C.)
- Department of Chemistry and Biochemistry, South Dakota State University, Brookings, SD 57007, USA
- Department of Pediatrics, Sanford School of Medicine, University of South Dakota, Vermillion, SD 57069, USA
| |
Collapse
|
10
|
Molstad DHH, Mattson AM, Begun DL, Westendorf JJ, Bradley EW. Hdac3 regulates bone modeling by suppressing osteoclast responsiveness to RANKL. J Biol Chem 2021; 295:17713-17723. [PMID: 33454009 DOI: 10.1074/jbc.ra120.013573] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 09/30/2020] [Indexed: 11/06/2022] Open
Abstract
Hdac3 is a lysine deacetylase that removes acetyl groups from histones and additional proteins. Although Hdac3 functions within mesenchymal lineage skeletal cells are defined, little is known about Hdac3 activities in bone-resorbing osteoclasts. In this study we conditionally deleted Hdac3 within Ctsk-expressing cells and examined the effects on bone modeling and osteoclast differentiation in mice. Hdac3 deficiency reduced femur and tibia periosteal circumference and increased cortical periosteal osteoclast number. Trabecular bone was likewise reduced and was accompanied by increased osteoclast number per trabecular bone surface. We previously showed that Hdac3 deacetylates the p65 subunit of the NF-κB transcriptional complex to decrease DNA-binding and transcriptional activity. Hdac3-deficient osteoclasts demonstrate increased K310 NF-κB acetylation and NF-κB transcriptional activity. Hdac3-deficient osteoclast lineage cells were hyper-responsive to RANKL and showed elevated ex vivo osteoclast number and size and enhanced bone resorption in pit formation assays. Osteoclast-directed Hdac3 deficiency decreased cortical and trabecular bone mass parameters, suggesting that Hdac3 regulates coupling of bone resorption and bone formation. We surveyed a panel of osteoclast-derived coupling factors and found that Hdac3 suppression diminished sphingosine-1-phosphate production. Osteoclast-derived sphingosine-1-phosphate acts in paracrine to promote bone mineralization. Mineralization of WT bone marrow stromal cells cultured with conditioned medium from Hdac3-deficient osteoclasts was markedly reduced. Expression of alkaline phosphatase, type 1a1 collagen, and osteocalcin was also suppressed, but no change in Runx2 expression was observed. Our results demonstrate that Hdac3 controls bone modeling by suppressing osteoclast lineage cell responsiveness to RANKL and coupling to bone formation.
Collapse
Affiliation(s)
- David H H Molstad
- Department of Orthopedics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Anna M Mattson
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Dana L Begun
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA
| | - Jennifer J Westendorf
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota, USA; Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota, USA
| | - Elizabeth W Bradley
- Department of Orthopedics, University of Minnesota, Minneapolis, Minnesota, USA; Stem Cell Institute, University of Minnesota, Minneapolis, Minnesota, USA.
| |
Collapse
|
11
|
Wang JS, Yoon SH, Wein MN. Role of histone deacetylases in bone development and skeletal disorders. Bone 2021; 143:115606. [PMID: 32829038 PMCID: PMC7770092 DOI: 10.1016/j.bone.2020.115606] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 08/11/2020] [Accepted: 08/15/2020] [Indexed: 02/08/2023]
Abstract
Bone cells must constantly respond to hormonal and mechanical cues to change gene expression programs. Of the myriad of epigenomic mechanisms used by cells to dynamically alter cell type-specific gene expression, histone acetylation and deacetylation has received intense focus over the past two decades. Histone deacetylases (HDACs) represent a large family of proteins with a conserved deacetylase domain first described to deacetylate lysine residues on histone tails. It is now appreciated that multiple classes of HDACs exist, some of which are clearly misnamed in that acetylated lysine residues on histone tails is not the major function of their deacetylase domain. Here, we will review the roles of proteins bearing deacetylase domains in bone cells, focusing on current genetic evidence for each individual HDAC gene. While class I HDACs are nuclear proteins whose primary role is to deacetylate histones, class IIa and class III HDACs serve other important cellular functions. Detailed knowledge of the roles of individual HDACs in bone development and remodeling will set the stage for future efforts to specifically target individual HDAC family members in the treatment of skeletal diseases such as osteoporosis.
Collapse
Affiliation(s)
- Jialiang S Wang
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sung-Hee Yoon
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Marc N Wein
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
12
|
Molstad DHH, Zars E, Norton A, Mansky KC, Westendorf JJ, Bradley EW. Hdac3 deletion in myeloid progenitor cells enhances bone healing in females and limits osteoclast fusion via Pmepa1. Sci Rep 2020; 10:21804. [PMID: 33311522 PMCID: PMC7733476 DOI: 10.1038/s41598-020-78364-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 11/06/2020] [Indexed: 12/18/2022] Open
Abstract
Previous studies examining the role of the histone deacetylase Hdac3 within myeloid cells demonstrated that Hdac3 promotes M2 activation and tissue healing in inflammatory conditions. Since myeloid lineage cells are required for proper bone formation and regeneration, in this study we examined the functions of Hdac3 during bone healing. Conditional deletion of Hdac3 within myeloid progenitors accelerates healing of cortical bone defects. Moreover, reduced osteoclast numbers within the defect site are correlated with Hdac3 suppression. Ex vivo osteoclastogenesis assays further demonstrate that Hdac3 deficiency limits osteoclastogenesis, the number of nuclei per cell and bone resorption, suggesting a defect in cell fusion. High throughput RNA sequencing identified the transmembrane protein Pmepa1 as a differentially expressed gene within osteoclast progenitor cells. Knockdown of Pmepa1 partially restores defects in osteoclastogenesis induced by Hdac3 deficiency. These results show that Hdac3 is required for optimal bone healing and osteoclast fusion, potentially via its regulation of Pmepa1 expression.
Collapse
Affiliation(s)
- David H H Molstad
- Department of Orthopedics, University of Minnesota, Elizabeth W. Bradley, 100 Church St. S.E., Minneapolis, MN, 55455, USA
| | - Elizabeth Zars
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andrew Norton
- Developmental and Surgical Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Kim C Mansky
- Developmental and Surgical Sciences, University of Minnesota, Minneapolis, MN, USA
| | - Jennifer J Westendorf
- Departments of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
- Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Elizabeth W Bradley
- Department of Orthopedics, University of Minnesota, Elizabeth W. Bradley, 100 Church St. S.E., Minneapolis, MN, 55455, USA.
- Stem Cell Institute, University of Minnesota, Minneapolis, MN, USA.
| |
Collapse
|
13
|
Cao LB, Leung CK, Law PWN, Lv Y, Ng CH, Liu HB, Lu G, Ma JL, Chan WY. Systemic changes in a mouse model of VCD-induced premature ovarian failure. Life Sci 2020; 262:118543. [PMID: 33038381 DOI: 10.1016/j.lfs.2020.118543] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023]
Abstract
AIMS Premature ovarian failure (POF) is a phenomenon in which the ovaries fail before the age of 40 years. Prior research has used a wide range of mouse models designed to reflect different causes of POF, including genetic factors, iatrogenic factors, and immune factors. The current study employed a mouse model of POF induced by 4-vinylcyclohexene diepoxide (VCD). VCD can specifically kill primordial and primary ovarian follicles, which destroys the follicular reserve and causes POF. The current study sought to specify and extend the applications of this model by examining the effect of timing and VCD dose and by exploring the effect of the model on systems outside of the ovaries. MATERIALS AND METHODS A VCD-induced mouse model of POF was constructed using established methods (VCD injected continuously at a concentration of 160 mg/kg for 15 days). Evidence for a graded effect of VCD was observed using a range of concentrations, and the best windows for examining VCD's effects on follicles and associated tissues were identified. KEY FINDINGS The mouse model used here successfully simulated two common complications of POF - emotional changes and decreased bone density. The model's application was then extended to examine the links between disease and intestinal microorganisms, and evidence was found linking POF to the reproductively relevant composition of the gut microbiota. SIGNIFICANCE These findings provide novel methodological guidance for future research, and they significantly extend the applications and scope of VCD-induced POF mouse models.
Collapse
Affiliation(s)
- Lian Bao Cao
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250001, China; CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China
| | - Chi Kwan Leung
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250001, China; CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China; SDIVF R&D Centre, Hong Kong Science and Technology Parks, Shatin, Hong Kong, China
| | - Patrick Wai-Nok Law
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China
| | - Yue Lv
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250001, China; CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China
| | - Cheuk-Hei Ng
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250001, China; CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China; SDIVF R&D Centre, Hong Kong Science and Technology Parks, Shatin, Hong Kong, China
| | - Hong Bin Liu
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250001, China; CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China; SDIVF R&D Centre, Hong Kong Science and Technology Parks, Shatin, Hong Kong, China
| | - Gang Lu
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China.
| | - Jin Long Ma
- Center for Reproductive Medicine, Shandong University, Jinan, Shandong 250001, China; CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China; SDIVF R&D Centre, Hong Kong Science and Technology Parks, Shatin, Hong Kong, China
| | - Wai Yee Chan
- CUHK-SDU Joint Laboratory on Reproductive Genetics, School of Biomedical Sciences, the Chinese University of Hong Kong, Hong Kong, China; National Research Center for Assisted Reproductive Technology and Reproductive Genetics, Jinan, Shandong 250001, China.
| |
Collapse
|
14
|
Zhou L, Wu H, Gao X, Zheng X, Chen H, Li H, Peng J, Liang W, Wang W, Qiu Z, Udduttula A, Wu K, Li L, Liu Y, Liu Y. Bone-Targeting Liposome-Encapsulated Salvianic Acid A Improves Nonunion Healing Through the Regulation of HDAC3-Mediated Endochondral Ossification. Drug Des Devel Ther 2020; 14:3519-3533. [PMID: 32982168 PMCID: PMC7502027 DOI: 10.2147/dddt.s263787] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/05/2020] [Indexed: 12/12/2022] Open
Abstract
AIM Nonunion is a major complication in fracture repair and remains a challenge in orthopaedics and trauma surgery. In this study, we aimed to evaluate the effectiveness of treatment of nonunion with a large radial defect using a bone-targeting liposome-encapsulated salvianic acid A (SAA-BTL)-incorporated collagen sponge and further elucidate whether the effects were closely related to histone deacetylase 3 (HDAC 3)-mediated endochondral ossification in nonunion healing process. METHODS Fifteen New Zealand female rabbits were randomly divided into three groups. Segmental radius critical size defects (15 mm) were created via surgery on both the forelimbs of the rabbits. The SAA-BTL/SAA/saline-incorporated collagen sponges were implanted into the defects in the three groups, respectively, for four weeks of treatment. X-ray imaging, micro-computed tomography (CT) analysis, histology, and immunofluorescence analysis (HDAC3, collagen II, VEGFA, and osteocalcin) were performed to determine the effects of the treatments. In addition, a short interfering RNA was applied to induce HDAC3 knockdown in the chondrogenic cell line ATDC5 to investigate the roles of HDAC3 and SAA intervention in endochondral ossification in nonunion healing. RESULTS X-ray imaging and micro-CT results revealed that SAA-BTL-incorporated collagen sponges significantly stimulated bone formation in the nonunion defect rabbit model. Furthermore, immunofluorescence double staining and histology analysis confirmed that SAA-BTL significantly increased the expression of P-HDAC3, collagen II, RUNX2, VEGFA, and osteocalcin in vivo; accelerated endochondral ossification turnover from cartilage to bone; and promoted long bone healing of nonunion defects. ATDC5 cells knocked down for HDAC3 showed significantly decreased expression of HDAC3, which resulted in reduced expression of chondrogenesis, osteogenesis, and angiogenesis biomarker genes (Sox9, Col10a1, VEGFA, RUNX2, and Col1a1), and increased expression of extracellular matrix degradation marker (MMP13). SAA treatment reversed these effects in the HDAC3 knockdown cell model. CONCLUSION SAA-BTL can improve nonunion healing through the regulation of HDAC3-mediated endochondral ossification.
Collapse
Affiliation(s)
- Limin Zhou
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Haojun Wu
- Department of Orthopaedics, Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong524001, People’s Republic of China
| | - Xiang Gao
- Department of Orthopaedics, Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong524001, People’s Republic of China
| | - Xiaoyan Zheng
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Hang Chen
- Department of Orthopaedics, Stem Cell Research and Cellular Therapy Center, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong524001, People’s Republic of China
| | - Hailong Li
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Jun Peng
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Weichong Liang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Wenxing Wang
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Zuocheng Qiu
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen518055, People’s Republic of China
| | - Anjaneyulu Udduttula
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen518055, People’s Republic of China
| | - Kefeng Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Lin Li
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou510515, Guangdong Province, People’s Republic of China
| | - Yuyu Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
| | - Yanzhi Liu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Department of Pharmacology, Guangdong Medical University, Zhanjiang, Guangdong524023, People’s Republic of China
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen518055, People’s Republic of China
| |
Collapse
|
15
|
Zhang L, Qi M, Chen J, Zhao J, Li L, Hu J, Jin Y, Liu W. Impaired autophagy triggered by HDAC9 in mesenchymal stem cells accelerates bone mass loss. Stem Cell Res Ther 2020; 11:269. [PMID: 32620134 PMCID: PMC7333327 DOI: 10.1186/s13287-020-01785-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 05/19/2020] [Accepted: 06/23/2020] [Indexed: 12/16/2022] Open
Abstract
Background Bone mass loss in aging is linked with imbalanced lineage differentiation of bone marrow mesenchymal stem cells (BMMSCs). Recent studies have proved that histone deacetylases (HDACs) are regarded as key regulators of bone remodeling. However, HDACs involve in regulating BMMSC bio-behaviors remain elusive. Here, we investigated the ability of HDAC9 on modulation of autophagy and its significance in lineage differentiation of BMMSCs. Methods The effects of HDAC9 on lineage differentiation of BMMSCs and autophagic signaling were assessed by various biochemical (western blot and ChIP assay), morphological (TEM and confocal microscopy), and micro-CT assays. Results Sixteen-month mice manifested obvious bone mass loss and marrow fat increase, accompanied with decreased osteogenic differentiation and increased adipogenic differentiation of BMMSCs. Further, the expression of HDAC9 elevated in bone and BMMSCs. Importantly, HDAC9 inhibitors recovered the lineage differentiation abnormality of 16-month BMMSCs and reduced p53 expression. Mechanistically, we revealed that HDAC9 regulated the autophagy of BMMSCs by controlling H3K9 acetylation in the promoters of the autophagic genes, ATG7, BECN1, and LC3a/b, which subsequently affected their lineage differentiation. Finally, HDAC9 inhibition improved endogenous BMMSC properties and promoted the bone mass recovery of 16-month mice. Conclusions Our data demonstrate that HDAC9 is a key regulator in a variety of bone mass by regulating autophagic activity in BMMSCs and thus a potential target of age-related bone loss treatment.
Collapse
Affiliation(s)
- Liqiang Zhang
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China.,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, Shaanxi, China.,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, Shaanxi, China
| | - Meng Qi
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Ji Chen
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Jiangdong Zhao
- The Key Laboratory of Aerospace Medicine, Ministry of Education, The Fourth Military Medical University, Xi'an, 710032, Shaanxi, China
| | - Liya Li
- Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, Shaanxi, China
| | - Jiachen Hu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, Shaanxi, China
| | - Yan Jin
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, Shaanxi, China. .,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, Shaanxi, China.
| | - Wenjia Liu
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, Precision Medicine Institute, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, China. .,State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi International Joint Research Center for Oral Diseases, Center for Tissue Engineering, School of Stomatology, The Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, Shaanxi, China. .,Xi'an Institute of Tissue Engineering and Regenerative Medicine, Xi'an, 710032, Shaanxi, China.
| |
Collapse
|
16
|
Louvet L, Leterme D, Delplace S, Miellot F, Marchandise P, Gauthier V, Hardouin P, Chauveau C, Ghali Mhenni O. Sirtuin 1 deficiency decreases bone mass and increases bone marrow adiposity in a mouse model of chronic energy deficiency. Bone 2020; 136:115361. [PMID: 32289519 DOI: 10.1016/j.bone.2020.115361] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 04/04/2020] [Accepted: 04/07/2020] [Indexed: 11/16/2022]
Abstract
Sirtuin of type 1 (Sirt1), a class III HDAC, is known to be involved in the regulation of differentiation of skeletal stem cells (SSCs) into osteoblasts and adipocytes. In caloric restriction, it has been shown that the expression and activity of Sirt1 is a tissue-dependent regulation. However, at present, no study has focused on the link between Sirt1, bone marrow adiposity (BMA) and osteoporosis related to anorexia nervosa (AN). Thus, the aims of this work were to (i) determine BMA and bone changes in a mouse model replicating the phenotypes of AN (separation-based anorexia model (SBA)); (ii) determine the expression of Sirt1 in bone marrow stromal cells (BMSCs) extracted from these mice and identify their differentiation capacities; (iii) study the effects of pharmacological activation and inhibition of Sirt1 on the osteoblastogenesis and adipogenesis of these cells and (iiii) delineate the molecular mechanism by which Sirt1 could regulate osteogenesis in an SBA model. Our results demonstrated that SBA protocol induces an increase in BMA and alteration of bone architecture. In addition, BMSCs from restricted mice present a down-regulation of Sirt1 which is accompanied by an increase in adipogenesis at expense of osteogenesis. After a 10-day organotypic culture, tibias from SBA mice displayed low levels of Sirt1 mRNA which are restored by resveratrol treatment. Interestingly, this recovery of Sirt1 levels also returned the BMA, BV/TV and Tb.Th in cultured tibias from SBA mice to normal levels. In contrast of down-regulation of Sirt1 expression induced by sirtinol treatment, stimulation of Sirt1 expression by resveratrol lead to a decrease in adipogenesis and increase in osteogenesis. Finally, to investigate the molecular mechanisms by which Sirt1 could regulate osteogenesis in the SBA model, the acetylation levels of Runx2 and Foxo1 transcription factors were determined. Our data show that this chronic energy deficiency in female mice causes a decrease in BMSC activity, resulting in critical changes to Runx2 and Foxo1 acetylation levels and thus to their activity. Altogether, these data suggest that Sirt1 could be considered as a potential therapeutic target in osteoporosis related to AN.
Collapse
Affiliation(s)
- Loïc Louvet
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Damien Leterme
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Séverine Delplace
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Flore Miellot
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Pierre Marchandise
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Véronique Gauthier
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Pierre Hardouin
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Christophe Chauveau
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France
| | - Olfa Ghali Mhenni
- Marrow Adiposity and Bone Lab (MABLab, ex-PMOI) ULR4490, Univ. Littoral Côte d'Opale F-62200 Boulogne-sur-Mer, Univ. Lille F-59000 Lille, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
17
|
Sharma AK, Roberts RL, Benson RD, Pierce JL, Yu K, Hamrick MW, McGee-Lawrence ME. The Senolytic Drug Navitoclax (ABT-263) Causes Trabecular Bone Loss and Impaired Osteoprogenitor Function in Aged Mice. Front Cell Dev Biol 2020; 8:354. [PMID: 32509782 PMCID: PMC7252306 DOI: 10.3389/fcell.2020.00354] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 04/21/2020] [Indexed: 12/02/2022] Open
Abstract
Senescence is a cellular defense mechanism that helps cells prevent acquired damage, but chronic senescence, as in aging, can contribute to the development of age-related tissue dysfunction and disease. Previous studies clearly show that removal of senescent cells can help prevent tissue dysfunction and extend healthspan during aging. Senescence increases with age in the skeletal system, and selective depletion of senescent cells or inhibition of their senescence-associated secretory phenotype (SASP) has been reported to maintain or improve bone mass in aged mice. This suggests that promoting the selective removal of senescent cells, via the use of senolytic agents, can be beneficial in the treatment of aging-related bone loss and osteoporosis. Navitoclax (also known as ABT-263) is a chemotherapeutic drug reported to effectively clear senescent hematopoietic stem cells, muscle stem cells, and mesenchymal stromal cells in previous studies, but its in vivo effects on bone mass had not yet been reported. Therefore, the purpose of this study was to assess the effects of short-term navitoclax treatment on bone mass and osteoprogenitor function in old mice. Aged (24 month old) male and female mice were treated with navitoclax (50 mg/kg body mass daily) for 2 weeks. Surprisingly, despite decreasing senescent cell burden, navitoclax treatment decreased trabecular bone volume fraction in aged female and male mice (−60.1% females, −45.6% males), and BMSC-derived osteoblasts from the navitoclax treated mice were impaired in their ability to produce a mineralized matrix (−88% females, −83% males). Moreover, in vitro administration of navitoclax decreased BMSC colony formation and calcified matrix production by aged BMSC-derived osteoblasts, similar to effects seen with the primary BMSC from the animals treated in vivo. Navitoclax also significantly increased metrics of cytotoxicity in both male and female osteogenic cultures (+1.0 to +11.3 fold). Taken together, these results suggest a potentially harmful effect of navitoclax on skeletal-lineage cells that should be explored further to definitively assess navitoclax’s potential (or risk) as a therapeutic agent for combatting age-related musculoskeletal dysfunction and bone loss.
Collapse
Affiliation(s)
- Anuj K Sharma
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Reginald D Benson
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Jessica L Pierce
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, United States.,Department of Orthopaedic Surgery, Augusta University, Augusta, GA, United States
| |
Collapse
|
18
|
Ding K, McGee-Lawrence ME, Kaiser H, Sharma AK, Pierce JL, Irsik DL, Bollag WB, Xu J, Zhong Q, Hill W, Shi XM, Fulzele S, Kennedy EJ, Elsalanty M, Hamrick MW, Isales CM. Picolinic acid, a tryptophan oxidation product, does not impact bone mineral density but increases marrow adiposity. Exp Gerontol 2020; 133:110885. [PMID: 32088397 PMCID: PMC7065047 DOI: 10.1016/j.exger.2020.110885] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2019] [Revised: 02/18/2020] [Accepted: 02/19/2020] [Indexed: 12/21/2022]
Abstract
Tryptophan is an essential amino acid catabolized initially to kynurenine (kyn), an immunomodulatory metabolite that we have previously shown to promote bone loss. Kyn levels increase with aging and have also been associated with neurodegenerative disorders. Picolinic acid (PA) is another tryptophan metabolite downstream of kyn. However, in contrast to kyn, PA is reported to be neuroprotective and further, to promote osteogenesis in vitro. Thus, we hypothesized that PA might be osteoprotective in vivo. In an IACUC-approved protocol, we fed PA to aged (23-month-old) C57BL/6 mice for eight weeks. In an effort to determine potential interactions of PA with dietary protein we also fed PA in a low-protein diet (8%). The mice were divided into four groups: Control (18% dietary protein), +PA (700 ppm); Low-protein (8%), +PA (700 ppm). The PA feedings had no impact on mouse weight, body composition or bone density. At sacrifice bone and stem cells were collected for analysis, including μCT and RT-qPCR. Addition of PA to the diet had no impact on trabecular bone parameters. However, marrow adiposity was significantly increased in PA-fed mice, and in bone marrow stromal cells isolated from these mice increases in the expression of the lipid storage genes, Plin1 and Cidec, were observed. Thus, as a downstream metabolite of kyn, PA no longer showed kyn's detrimental effects on bone but instead appears to impact energy balance.
Collapse
Affiliation(s)
- Kehong Ding
- Center for Healthy Aging, Augusta University, United States of America; Department of Neuroscience and Regenerative Medicine, Augusta University, United States of America
| | - Meghan E McGee-Lawrence
- Center for Healthy Aging, Augusta University, United States of America; Department of Orthopaedic Surgery, Augusta University, United States of America; Department of Cellular Biology and Anatomy, Augusta University, United States of America
| | - Helen Kaiser
- Department of Cellular Biology and Anatomy, Augusta University, United States of America
| | - Anuj K Sharma
- Department of Cellular Biology and Anatomy, Augusta University, United States of America
| | - Jessica L Pierce
- Department of Cellular Biology and Anatomy, Augusta University, United States of America
| | - Debra L Irsik
- Department of Neuroscience and Regenerative Medicine, Augusta University, United States of America
| | - Wendy B Bollag
- Center for Healthy Aging, Augusta University, United States of America; Department of Medicine, Augusta University, United States of America; Department of Physiology, Augusta University, United States of America; Charlie Norwood VA Medical Center, Augusta, GA 30912, United States of America
| | - Jianrui Xu
- Department of Neuroscience and Regenerative Medicine, Augusta University, United States of America
| | - Qing Zhong
- Center for Healthy Aging, Augusta University, United States of America; Department of Neuroscience and Regenerative Medicine, Augusta University, United States of America
| | - William Hill
- Center for Healthy Aging, Augusta University, United States of America; Department of Pathology and Laboratory Medicine, Medical University of South Carolina, United States of America; Ralph H Johnson Veterans Affairs Medical Center, Charleston, SC 29403, United States of America
| | - Xing-Ming Shi
- Center for Healthy Aging, Augusta University, United States of America; Department of Neuroscience and Regenerative Medicine, Augusta University, United States of America; Department of Orthopaedic Surgery, Augusta University, United States of America
| | - Sadanand Fulzele
- Center for Healthy Aging, Augusta University, United States of America; Department of Orthopaedic Surgery, Augusta University, United States of America
| | - Eileen J Kennedy
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA 30602, United States of America
| | - Mohammed Elsalanty
- Center for Healthy Aging, Augusta University, United States of America; Department of Oral Biology, Augusta University, United States of America
| | - Mark W Hamrick
- Center for Healthy Aging, Augusta University, United States of America; Department of Orthopaedic Surgery, Augusta University, United States of America; Department of Cellular Biology and Anatomy, Augusta University, United States of America
| | - Carlos M Isales
- Center for Healthy Aging, Augusta University, United States of America; Department of Medicine, Augusta University, United States of America; Department of Neuroscience and Regenerative Medicine, Augusta University, United States of America; Department of Orthopaedic Surgery, Augusta University, United States of America; Department of Cellular Biology and Anatomy, Augusta University, United States of America.
| |
Collapse
|
19
|
Pierce JL, Roberts RL, Yu K, Kendall RK, Kaiser H, Davis C, Johnson MH, Hill WD, Isales CM, Bollag WB, Hamrick MW, McGee-Lawrence ME. Kynurenine suppresses osteoblastic cell energetics in vitro and osteoblast numbers in vivo. Exp Gerontol 2020; 130:110818. [PMID: 31862422 PMCID: PMC7003726 DOI: 10.1016/j.exger.2019.110818] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/13/2019] [Accepted: 12/16/2019] [Indexed: 12/11/2022]
Abstract
Aging is a progressive process associated with declining tissue function over time. Kynurenine, an oxidized metabolite of the essential amino acid tryptophan that increases in abundance with age, drives cellular processes of aging and dysfunction in many tissues, and recent work has focused on understanding the pathways involved in the harmful effects of kynurenine on bone. In this study, we sought to investigate the effects of controlled kynurenine administration on osteoblast bioenergetics, in vivo osteoblast abundance, and marrow fat accumulation. Additionally, as an extension of earlier studies with dietary administration of kynurenine, we investigated the effects of kynurenine on Hdac3 and NCoR1 expression and enzymatic deacetylase activity as potential mechanistic contributors to the effects of kynurenine on osteoblasts. Kynurenine administration suppressed cellular metabolism in osteoblasts at least in part through impaired mitochondrial respiration, and suppressed osteoblastic numbers in vivo with no concurrent effects on marrow adiposity. Deleterious effects of kynurenine treatment on osteoblasts were more pronounced in female models as compared to males. However, kynurenine treatment did not inhibit Hdac3's enzymatic deacetylase activity nor its repression of downstream glucocorticoid signaling. As such, future work will be necessary to determine the mechanisms by which increased kynurenine contributes to aging bone bioenergetics. The current study provides novel further support for the idea that kynurenine contributes to impaired osteoblastic function, and suggests that impaired matrix production by kynurenine-affected osteoblasts is attributed in part to impaired osteoblastic bioenergetics. As circulating kynurenine levels in increase with age, and human bone density inversely correlates with the serum kynurenine to tryptophan ratio, these mechanisms may have important relevance in the etiology and pathogenesis of osteoporosis in humans.
Collapse
Affiliation(s)
- Jessica L Pierce
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Rachel L Roberts
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Riley K Kendall
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Helen Kaiser
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Colleen Davis
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Biological Sciences, Augusta University, Augusta, GA, USA
| | - Maribeth H Johnson
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA
| | - William D Hill
- Department of Pathology and Laboratory Medicine, College of Medicine, Medical University of South Carolina, Charleston, SC, USA
| | - Carlos M Isales
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, USA; Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA; Department of Medicine, Division of Endocrinology, Diabetes and Metabolism, Augusta University, Augusta, GA, USA
| | - Wendy B Bollag
- Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA; Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA, USA; Charlie Norwood Veterans' Affairs Medical Center, Augusta, GA, USA
| | - Mark W Hamrick
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopaedic Surgery, Augusta University, Augusta, GA, USA.
| |
Collapse
|
20
|
Emmett MJ, Lazar MA. Integrative regulation of physiology by histone deacetylase 3. Nat Rev Mol Cell Biol 2019; 20:102-115. [PMID: 30390028 DOI: 10.1038/s41580-018-0076-0] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Cell-type-specific gene expression is physiologically modulated by the binding of transcription factors to genomic enhancer sequences, to which chromatin modifiers such as histone deacetylases (HDACs) are recruited. Drugs that inhibit HDACs are in clinical use but lack specificity. HDAC3 is a stoichiometric component of nuclear receptor co-repressor complexes whose enzymatic activity depends on this interaction. HDAC3 is required for many aspects of mammalian development and physiology, for example, for controlling metabolism and circadian rhythms. In this Review, we discuss the mechanisms by which HDAC3 regulates cell type-specific enhancers, the structure of HDAC3 and its function as part of nuclear receptor co-repressors, its enzymatic activity and its post-translational modifications. We then discuss the plethora of tissue-specific physiological functions of HDAC3.
Collapse
Affiliation(s)
- Matthew J Emmett
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Mitchell A Lazar
- Institute for Diabetes, Obesity, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA. .,Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA.
| |
Collapse
|
21
|
Shreya S, Malavika D, Priya VR, Selvamurugan N. Regulation of Histone Deacetylases by MicroRNAs in Bone. Curr Protein Pept Sci 2019; 20:356-367. [PMID: 30381072 DOI: 10.2174/1389203720666181031143129] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2018] [Revised: 10/15/2018] [Accepted: 10/19/2018] [Indexed: 02/08/2023]
Abstract
Formation of new bone by osteoblasts is mediated via the activation of signaling pathways, such as TGF-β, BMP, and Wnt. A number of transcription factors participate in the signaling cascades that are tightly regulated by other regulatory factors. Histone deacetylases (HDACs) are one such class of regulatory factors that play an essential role in influencing chromatin architecture and regulate the expression of the genes that play a role in osteoblast differentiation by the mechanism of deacetylation. Four classes of HDACs have been identified namely, class I, class II A, class II B, class III and class IV. MicroRNAs (miRNAs) are small fragments of non-coding RNAs typically 19-25 nucleotides long that target mRNAs to upregulate or downregulate gene expression at a post-transcriptional level. A number of miRNAs that target HDACs in bone have been recently reported. Hence, in this review, we elaborate on the various miRNAs that target the different classes of HDACs and impact of the same on osteogenesis.
Collapse
Affiliation(s)
- S Shreya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - D Malavika
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - V Raj Priya
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| | - N Selvamurugan
- Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur 603 203, Tamil Nadu, India
| |
Collapse
|
22
|
Berent TE, Dorschner JM, Craig TA, Drake MT, Westendorf JJ, Kumar R. Lung tumor cells inhibit bone mineralization and osteoblast activity. Biochem Biophys Res Commun 2019; 519:566-571. [PMID: 31537378 DOI: 10.1016/j.bbrc.2019.09.045] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 09/11/2019] [Indexed: 11/26/2022]
Abstract
Patients with non-small cell lung cancer (NSLC) often develop skeletal complications and fractures. To understand mechanisms of bone loss, we developed a murine model of non-metastatic NSLC. Decreased bone mineral density, trabecular thickness and mineralization, without an increase in bone resorption, were observed in vivo in mice injected with Lewis lung adenocarcinoma (LLC1) cells in the absence of tumor cell metastases. A decrease in trabecular bone mineral density was observed in mice injected with cell-free LLC1 CM. Plasma osteoblast biomarkers and PTH-related peptide (PTHrP) were reduced, and parathyroid hormone (PTH), 1,25-dihydroxyvitamin D, calcium and phosphate concentrations were normal in tumor-bearing mice. LLC1 cell conditioned medium (CM) inhibited alkaline phosphatase activity, osteoblast mineralization, and expression of Alpl and Ocn/Bglap mRNA in MC3T3 osteoblast cultures, whereas non-CM or CM from NIH/3T3 fibroblasts did not induce similar changes. LLC1 CM reduced Wnt3a-stimulated Tcf/Lef reporter plasmid activity and Wnt5A, Tcf1 and Lef1 mRNA expression in MC3T3 cells. Although concentrations of the Wnt inhibitor, DKK2, were increased in LLC1 CM compared to non-CM, depletion of DKK2 from LLC1 CM did not completely restore Wnt3a activity in MC3T3 cultures, and recombinant DKK2 failed to inhibit osteoblast mineralization. The data indicate that in a model of lung adenocarcinoma without bone metastases, tumor cells elaborate a secreted factor(s) that reduces bone mass, bone formation and osteoblast Wnt signaling without increases in bone resorption or calcium-regulating hormone concentrations. The factor(s) mediating this inhibition of osteoblast mineralization require further characterization.
Collapse
Affiliation(s)
- Taylor E Berent
- Department of Medicine, Divisions of Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jessica M Dorschner
- Department of Medicine, Divisions of Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Theodore A Craig
- Department of Medicine, Divisions of Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Matthew T Drake
- Department of Medicine, Divisions of Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
| | - Rajiv Kumar
- Department of Medicine, Divisions of Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Nephrology and Hypertension and Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Endocrinology, Diabetes, Metabolism and Nutrition, Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA; Department of Biochemistry and Molecular Biology, Department of Orthopedic Surgery, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA.
| |
Collapse
|
23
|
Pierce JL, Ding KH, Xu J, Sharma AK, Yu K, Del Mazo Arbona N, Rodriguez-Santos Z, Bernard P, Bollag WB, Johnson MH, Hamrick MW, Begun DL, Shi XM, Isales CM, McGee-Lawrence ME. The glucocorticoid receptor in osteoprogenitors regulates bone mass and marrow fat. J Endocrinol 2019; 243:JOE-19-0230.R1. [PMID: 31370004 PMCID: PMC6938567 DOI: 10.1530/joe-19-0230] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Accepted: 07/31/2019] [Indexed: 12/18/2022]
Abstract
Excess fat within bone marrow is associated with lower bone density. Metabolic stressors such as chronic caloric restriction (CR) can exacerbate marrow adiposity, and increased glucocorticoid signaling and adrenergic signaling are implicated in this phenotype. The current study tested the role of glucocorticoid signaling in CR-induced stress by conditionally deleting the glucocorticoid receptor (GR) in bone marrow osteoprogenitors (Osx1-Cre) of mice subjected to CR and ad libitum diets. Conditional knockout of the GR (GR-CKO) reduced cortical and trabecular bone mass as compared to wildtype (WT) mice under both ad libitum and CR conditions. No interaction was detected between genotype and diet, suggesting that the GR is not required for CR-induced skeletal changes. The lower bone mass in GR-CKO mice, and the further suppression of bone by CR, resulted from suppressed bone formation. Interestingly, treatment with the -adrenergic receptor antagonist propranolol mildly but selectively improved metrics of cortical bone mass in GR-CKO mice during CR, suggesting interaction between adrenergic and glucocorticoid signaling pathways that affects cortical bone. GR-CKO mice dramatically increased marrow fat under both ad libitum and CR-fed conditions, and surprisingly propranolol treatment was unable to rescue CR-induced marrow fat in either WT or GR-CKO mice. Additionally, serum corticosterone levels were selectively elevated in GR-CKO mice with CR, suggesting the possibility of bone-hypothalamus-pituitary-adrenal crosstalk during metabolic stress. This work highlights the complexities of glucocorticoid and β-adrenergic signaling in stress-induced changes in bone mass, and the importance of GR function in suppressing marrow adipogenesis while maintaining healthy bone mass.
Collapse
Affiliation(s)
- Jessica L Pierce
- J Pierce, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Ke-Hong Ding
- K Ding, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Jianrui Xu
- J Xu, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Anuj K Sharma
- A Sharma, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Kanglun Yu
- K Yu, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | | | | | - Paul Bernard
- P Bernard, Pediatric Endocrine Specialists of Georgia, Pediatric Endocrine Specialists of Georgia, Duluth, United States
| | - Wendy B Bollag
- W Bollag, Department of Physiology, Medical College of Georgia, Augusta, GA 30912, United States
| | - Maribeth H Johnson
- M Johnson, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Mark W Hamrick
- M Hamrick, Cellular Biology and Anatomy, Augusta University, Augusta, United States
| | - Dana L Begun
- D Begun, Department of Orthopedic Surgery, Mayo Clinic, Rochester, United States
| | - Xing M Shi
- X Ming Shi, Neuroscience and Regenerative Medicine, Augusta University, Augusta, United States
| | - Carlos M Isales
- C Isales, Neuroscience and Regenerative Medicine, Augusta University, Augusta, 30912, United States
| | | |
Collapse
|
24
|
Grote C, Reinhardt D, Zhang M, Wang J. Regulatory mechanisms and clinical manifestations of musculoskeletal aging. J Orthop Res 2019; 37:1475-1488. [PMID: 30919498 PMCID: PMC9202363 DOI: 10.1002/jor.24292] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 03/13/2019] [Indexed: 02/04/2023]
Abstract
Aging is the strongest risk factor for degenerative bone and joint diseases. Clinical therapies for age-related musculoskeletal disorders face significant challenges as their pathogenic mechanisms remain largely unclear. This review article focuses on the recent advances in the understanding of regulatory mechanisms of musculoskeletal aging and their clinical relevance. We begin with the prevalence and socioeconomic impacts of major age-related musculoskeletal disorders such as sarcopenia, osteoporosis, osteoarthritis, and degenerative tendinopathy. The current understanding of responsible biological mechanisms involved in general aging is then summarized. Proposed molecular, cellular, and biomechanical mechanisms relevant to the clinical manifestations of aging in the musculoskeletal system are discussed in detail, with a focus on the disorders affecting muscle, bone, articular cartilage, and tendon. Although musculoskeletal aging processes share many common pathways with the aging of other body systems, unique molecular and cellular mechanisms may be involved in the aging processes of musculoskeletal tissues. Advancements in the understanding of regulatory mechanisms of musculoskeletal aging may promote the development of novel treatments for age-related musculoskeletal disorders. Finally, future research directions for major musculoskeletal tissues including functional interaction between the tissues and their clinical relevance to age-related musculoskeletal disorders are highlighted in the Future Prospects section. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:1475-1488, 2019.
Collapse
Affiliation(s)
- Caleb Grote
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Daniel Reinhardt
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Mingcai Zhang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
| | - Jinxi Wang
- Harrington Laboratory for Molecular Orthopedics, Department of Orthopedic Surgery, University of Kansas Medical Center, Kansas City, Kansas, USA
- Department of Biochemistry & Molecular Biology, University of Kansas Medical Center, Kansas City, Kansas, USA
| |
Collapse
|
25
|
Lawlor L, Yang XB. Harnessing the HDAC-histone deacetylase enzymes, inhibitors and how these can be utilised in tissue engineering. Int J Oral Sci 2019; 11:20. [PMID: 31201303 PMCID: PMC6572769 DOI: 10.1038/s41368-019-0053-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 05/02/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
There are large knowledge gaps regarding how to control stem cells growth and differentiation. The limitations of currently available technologies, such as growth factors and/or gene therapies has led to the search of alternatives. We explore here how a cell's epigenome influences determination of cell type, and potential applications in tissue engineering. A prevalent epigenetic modification is the acetylation of DNA core histone proteins. Acetylation levels heavily influence gene transcription. Histone deacetylase (HDAC) enzymes can remove these acetyl groups, leading to the formation of a condensed and more transcriptionally silenced chromatin. Histone deacetylase inhibitors (HDACis) can inhibit these enzymes, resulting in the increased acetylation of histones, thereby affecting gene expression. There is strong evidence to suggest that HDACis can be utilised in stem cell therapies and tissue engineering, potentially providing novel tools to control stem cell fate. This review introduces the structure/function of HDAC enzymes and their links to different tissue types (specifically bone, cardiac, neural tissues), including the history, current status and future perspectives of using HDACis for stem cell research and tissue engineering, with particular attention paid to how different HDAC isoforms may be integral to this field.
Collapse
Affiliation(s)
- Liam Lawlor
- Department of Oral Biology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, UK
- Doctoral Training Centre in Tissue Engineering and Regenerative Medicine, Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, UK
| | - Xuebin B Yang
- Department of Oral Biology, University of Leeds, Wellcome Trust Brenner Building, St. James's University Hospital, Leeds, LS9 7TF, UK.
- Doctoral Training Centre in Tissue Engineering and Regenerative Medicine, Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, UK.
| |
Collapse
|
26
|
Ehnert S, Aspera-Werz RH, Ruoß M, Dooley S, Hengstler JG, Nadalin S, Relja B, Badke A, Nussler AK. Hepatic Osteodystrophy-Molecular Mechanisms Proposed to Favor Its Development. Int J Mol Sci 2019; 20:2555. [PMID: 31137669 PMCID: PMC6566554 DOI: 10.3390/ijms20102555] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 05/14/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023] Open
Abstract
Almost all patients with chronic liver diseases (CLD) show altered bone metabolism. Depending on the etiology, this manifests in a severe osteoporosis in up to 75% of the affected patients. Due to high prevalence, the generic term hepatic osteodystrophy (HOD) evolved, describing altered bone metabolism, decreased bone mineral density, and deterioration of bone structure in patients with CLD. Once developed, HOD is difficult to treat and increases the risk of fragility fractures. Existing fractures affect the quality of life and, more importantly, long-term prognosis of these patients, which presents with increased mortality. Thus, special care is required to support the healing process. However, for early diagnosis (reduce fracture risk) and development of adequate treatment strategies (support healing of existing fractures), it is essential to understand the underlying mechanisms that link disturbed liver function with this bone phenotype. In the present review, we summarize proposed molecular mechanisms favoring the development of HOD and compromising the healing of associated fractures, including alterations in vitamin D metabolism and action, disbalances in transforming growth factor beta (TGF-β) and bone morphogenetic protein (BMP) signaling with histone deacetylases (HDACs) as secondary regulators, as well as alterations in the receptor activator of nuclear factor kappa B ligand (RANKL)-osteoprotegerin (OPG) system mediated by sclerostin. Based on these mechanisms, we give an overview on the limitations of early diagnosis of HOD with established serum markers.
Collapse
Affiliation(s)
- Sabrina Ehnert
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Romina H Aspera-Werz
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Marc Ruoß
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Steven Dooley
- Department of Medicine II, Molecular Hepatology, Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany.
| | - Jan G Hengstler
- IfADo-Leibniz Research Centre for Working Environment and Human Factors, Technical University Dortmund, 44139 Dortmund, Germany.
| | - Silvio Nadalin
- Department of General, Visceral and Transplant Surgery, University Hospital Tuebingen, 72076 Tuebingen, Germany.
| | - Borna Relja
- Department of Trauma, Hand and Reconstructive Surgery, University Hospital Frankfurt, Goethe University, 60590 Frankfurt, Germany.
| | - Andreas Badke
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| | - Andreas K Nussler
- Siegfried Weller Research Institute, Department of Trauma and Reconstructive Surgery, Eberhard Karls University Tuebingen, BG Trauma Center Tuebingen, 72076 Tuebingen, Germany.
| |
Collapse
|
27
|
Sun N, Uda Y, Azab E, Kochen A, Santos RNCE, Shi C, Kobayashi T, Wein MN, Divieti Pajevic P. Effects of histone deacetylase inhibitor Scriptaid and parathyroid hormone on osteocyte functions and metabolism. J Biol Chem 2019; 294:9722-9733. [PMID: 31068415 DOI: 10.1074/jbc.ra118.007312] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 05/02/2019] [Indexed: 01/02/2023] Open
Abstract
Bone is a highly metabolic organ that undergoes continuous remodeling to maintain its structural integrity. During development, bones, in particular osteoblasts, rely on glucose uptake. However, the role of glucose metabolism in osteocytes is unknown. Osteocytes are terminally differentiated osteoblasts orchestrating bone modeling and remodeling. In these cells, parathyroid hormone (PTH) suppresses Sost/sclerostin expression (a potent inhibitor of bone formation) by promoting nuclear translocation of class IIa histone deacetylase (HDAC) 4 and 5 and the repression of myocyte enhancer factor 2 (MEF2) type C. Recently, Scriptaid, an HDAC complex co-repressor inhibitor, has been shown to induce MEF2 activation and exercise-like adaptation in mice. In muscles, Scriptaid disrupts the HDAC4/5 co-repressor complex, increases MEF2C function, and promotes cell respiration. We hypothesized that Scriptaid, by affecting HDAC4/5 localization and MEF2C activation, might affect osteocyte functions. Treatment of the osteocytic Ocy454-12H cells with Scriptaid increased metabolic gene expression, cell respiration, and glucose uptake. Similar effects were also seen upon treatment with PTH, suggesting that both Scriptaid and PTH can promote osteocyte metabolism. Similar to PTH, Scriptaid potently suppressed Sost expression. Silencing of HDAC5 in Ocy454-12H cells abolished Sost suppression but not glucose transporter type 4 (Glut4) up-regulation induced by Scriptaid. These results demonstrate that Scriptaid increases osteocyte respiration and glucose uptake by mechanisms independent of HDAC complex inhibition. In osteocytes, Scriptaid, similar to PTH, increases binding of HDAC5 to Mef2c with suppression of Sost but only partially increases receptor activator of NF-κB ligand (Rankl) expression, suggesting a potential bone anabolic effect.
Collapse
Affiliation(s)
- Ningyuan Sun
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118
| | - Yuhei Uda
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118
| | - Ehab Azab
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118
| | - Alejandro Kochen
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118
| | - Roberto Nunes Campos E Santos
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118
| | - Chao Shi
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118.,the Department of Orthopedics, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China, and
| | - Tokio Kobayashi
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118
| | - Marc N Wein
- the Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114
| | - Paola Divieti Pajevic
- From the Department of Molecular and Cell Biology, Henry M. Goldman School of Dental Medicine, Boston University, Boston, Massachusetts 02118,
| |
Collapse
|
28
|
Behera J, Kelly KE, Voor MJ, Metreveli N, Tyagi SC, Tyagi N. Hydrogen Sulfide Promotes Bone Homeostasis by Balancing Inflammatory Cytokine Signaling in CBS-Deficient Mice through an Epigenetic Mechanism. Sci Rep 2018; 8:15226. [PMID: 30323246 PMCID: PMC6189133 DOI: 10.1038/s41598-018-33149-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 09/18/2018] [Indexed: 01/28/2023] Open
Abstract
Previously, we have shown hyperhomocysteinemia (HHcy) to have a detrimental effect on bone remodeling, which is associated with osteoporosis. During transsulfuration, Hcy is metabolized into hydrogen sulfide (H2S), a gasotransmitter molecule known to regulate bone formation. Therefore, in the present study, we examined whether H2S ameliorates HHcy induced epigenetic and molecular alterations leading to osteoporotic bone loss. To test this mechanism, we employed cystathionine-beta-synthase heterozygote knockout mice, fed with a methionine rich diet (CBS+/− +Met), supplemented with H2S-donor NaHS for 8 weeks. Treatment with NaHS, normalizes plasma H2S, and completely prevents trabecular bone loss in CBS+/− mice. Our data showed that HHcy caused inhibition of HDAC3 activity and subsequent inflammation by imbalancing redox homeostasis. The mechanistic study revealed that inflammatory cytokines (IL-6, TNF-α) are transcriptionally activated by an acetylated lysine residue in histone (H3K27ac) of chromatin by binding to its promoter and subsequently regulating gene expression. A blockade of HDAC3 inhibition in CBS+/− mice by HDAC activator ITSA-1, led to the remodeling of histone landscapes in the genome and thereby attenuated histone acetylation-dependent inflammatory signaling. We also confirmed that RUNX2 was sulfhydrated by administration of NaHS. Collectively, restoration of H2S may provide a novel treatment for CBS-deficiency induced metabolic osteoporosis.
Collapse
Affiliation(s)
- Jyotirmaya Behera
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Kimberly E Kelly
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Michael J Voor
- Department of Orthopaedic Surgery, School of Medicine, University of Louisville, Louisville, KY, 40292, USA.,Department of Bioengineering, Speed School of Engineering, University of Louisville, Louisville, KY, 40292, USA
| | - Naira Metreveli
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Suresh C Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA
| | - Neetu Tyagi
- Bone Biology Laboratory, Department of Physiology, School of Medicine, University of Louisville, Louisville, KY, 40292, USA.
| |
Collapse
|
29
|
Araya HF, Sepulveda H, Lizama CO, Vega OA, Jerez S, Briceño PF, Thaler R, Riester SM, Antonelli M, Salazar-Onfray F, Rodríguez JP, Moreno RD, Montecino M, Charbonneau M, Dubois CM, Stein GS, van Wijnen AJ, Galindo MA. Expression of the ectodomain-releasing protease ADAM17 is directly regulated by the osteosarcoma and bone-related transcription factor RUNX2. J Cell Biochem 2018; 119:8204-8219. [PMID: 29923217 DOI: 10.1002/jcb.26832] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 03/06/2018] [Indexed: 01/04/2023]
Abstract
Osteoblast differentiation is controlled by transcription factor RUNX2 which temporally activates or represses several bone-related genes, including those encoding extracellular matrix proteins or factors that control cell-cell, and cell-matrix interactions. Cell-cell communication in the many skeletal pericellular micro-niches is critical for bone development and involves paracrine secretion of growth factors and morphogens. This paracrine signaling is in part regulated by "A Disintegrin And Metalloproteinase" (ADAM) proteins. These cell membrane-associated metalloproteinases support proteolytic release ("shedding") of protein ectodomains residing at the cell surface. We analyzed microarray and RNA-sequencing data for Adam genes and show that Adam17, Adam10, and Adam9 are stimulated during BMP2 mediated induction of osteogenic differentiation and are robustly expressed in human osteoblastic cells. ADAM17, which was initially identified as a tumor necrosis factor alpha (TNFα) converting enzyme also called (TACE), regulates TNFα-signaling pathway, which inhibits osteoblast differentiation. We demonstrate that Adam17 expression is suppressed by RUNX2 during osteoblast differentiation through the proximal Adam17 promoter region (-0.4 kb) containing two functional RUNX2 binding motifs. Adam17 downregulation during osteoblast differentiation is paralleled by increased RUNX2 expression, cytoplasmic-nuclear translocation and enhanced binding to the Adam17 proximal promoter. Forced expression of Adam17 reduces Runx2 and Alpl expression, indicating that Adam17 may negatively modulate osteoblast differentiation. These findings suggest a novel regulatory mechanism involving a reciprocal Runx2-Adam17 negative feedback loop to regulate progression through osteoblast differentiation. Our results suggest that RUNX2 may control paracrine signaling through regulation of ectodomain shedding at the cell surface of osteoblasts by directly suppressing Adam17 expression.
Collapse
Affiliation(s)
- Héctor F Araya
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Hugo Sepulveda
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Carlos O Lizama
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Oscar A Vega
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sofia Jerez
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Pedro F Briceño
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Marcelo Antonelli
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Flavio Salazar-Onfray
- Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile.,Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Juan Pablo Rodríguez
- Instituto de Nutrición y Tecnología de los Alimentos (INTA), University of Chile, Santiago, Chile
| | - Ricardo D Moreno
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Martin Montecino
- Center for Biomedical Research, Faculty of Biological Sciences and Faculty of Medicine, FONDAP Center for Genome Regulation, Universidad Andres Bello, Santiago, Chile
| | - Martine Charbonneau
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Claire M Dubois
- Immunology Division, Department of Pediatrics, Faculty of Medicine and Health Sciences, University of Sherbrooke, Sherbrooke, Quebec, Canada
| | - Gary S Stein
- Department of Biochemistry and University of Vermont Cancer Center, The Robert Larner MD College of Medicine, University of Vermont, Burlington, Vermont
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| | - Mario A Galindo
- Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.,Millennium Institute on Immunology and Immunotherapy, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
30
|
Dudakovic A, Camilleri ET, Paradise CR, Samsonraj RM, Gluscevic M, Paggi CA, Begun DL, Khani F, Pichurin O, Ahmed FS, Elsayed R, Elsalanty M, McGee-Lawrence ME, Karperien M, Riester SM, Thaler R, Westendorf JJ, van Wijnen AJ. Enhancer of zeste homolog 2 ( Ezh2) controls bone formation and cell cycle progression during osteogenesis in mice. J Biol Chem 2018; 293:12894-12907. [PMID: 29899112 DOI: 10.1074/jbc.ra118.002983] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Revised: 06/12/2018] [Indexed: 12/25/2022] Open
Abstract
Epigenetic mechanisms control skeletal development and osteoblast differentiation. Pharmacological inhibition of the histone 3 Lys-27 (H3K27) methyltransferase enhancer of zeste homolog 2 (EZH2) in WT mice enhances osteogenesis and stimulates bone formation. However, conditional genetic loss of Ezh2 early in the mesenchymal lineage (i.e. through excision via Prrx1 promoter-driven Cre) causes skeletal abnormalities due to patterning defects. Here, we addressed the key question of whether Ezh2 controls osteoblastogenesis at later developmental stages beyond patterning. We show that Ezh2 loss in committed pre-osteoblasts by Cre expression via the osterix/Sp7 promoter yields phenotypically normal mice. These Ezh2 conditional knock-out mice (Ezh2 cKO) have normal skull bones, clavicles, and long bones but exhibit increased bone marrow adiposity and reduced male body weight. Remarkably, in vivo Ezh2 loss results in a low trabecular bone phenotype in young mice as measured by micro-computed tomography and histomorphometry. Thus, Ezh2 affects bone formation stage-dependently. We further show that Ezh2 loss in bone marrow-derived mesenchymal cells suppresses osteogenic differentiation and impedes cell cycle progression as reflected by decreased metabolic activity, reduced cell numbers, and changes in cell cycle distribution and in expression of cell cycle markers. RNA-Seq analysis of Ezh2 cKO calvaria revealed that the cyclin-dependent kinase inhibitor Cdkn2a is the most prominent cell cycle target of Ezh2 Hence, genetic loss of Ezh2 in mouse pre-osteoblasts inhibits osteogenesis in part by inducing cell cycle changes. Our results suggest that Ezh2 serves a bifunctional role during bone formation by suppressing osteogenic lineage commitment while simultaneously facilitating proliferative expansion of osteoprogenitor cells.
Collapse
Affiliation(s)
- Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Emily T Camilleri
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Christopher R Paradise
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota 55905; Center for Regenerative Medicine, Mayo Clinic, Rochester, Minnesota 55905
| | | | - Martina Gluscevic
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, Minnesota 55905
| | - Carlo Alberto Paggi
- Department of Developmental BioEngineering, University of Twente, 7522 NB Enschede, Netherlands
| | - Dana L Begun
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Farzaneh Khani
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Oksana Pichurin
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Farah S Ahmed
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Ranya Elsayed
- Department of Oral Biology, Augusta University, Augusta, Georgia 30912
| | | | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia 30912; Department of Orthopedic Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | - Marcel Karperien
- Department of Developmental BioEngineering, University of Twente, 7522 NB Enschede, Netherlands
| | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Roman Thaler
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota 55905; Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota 55905.
| |
Collapse
|
31
|
Bollag WB, Choudhary V, Zhong Q, Ding KH, Xu J, Elsayed R, Yu K, Su Y, Bailey LJ, Shi XM, Elsalanty M, Johnson MH, McGee-Lawrence ME, Isales CM. Deletion of protein kinase D1 in osteoprogenitor cells results in decreased osteogenesis in vitro and reduced bone mineral density in vivo. Mol Cell Endocrinol 2018; 461:22-31. [PMID: 28811183 PMCID: PMC5756499 DOI: 10.1016/j.mce.2017.08.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2017] [Revised: 07/14/2017] [Accepted: 08/10/2017] [Indexed: 01/08/2023]
Abstract
Protein kinase D1 (PRKD1) is thought to play a role in a number of cellular functions, including proliferation and differentiation. We hypothesized that PRKD1 in bone marrow-derived mesenchymal stem cells (BMMSC) could modulate osteogenesis. In BMMSCs from floxed PRKD1 mice, PRKD1 ablation with adenovirus-mediated Cre-recombinase expression inhibited BMMSC differentiation in vitro. In 3- and 6-month-old conditional knockout mice (cKO), in which PRKD1 was ablated in osteoprogenitor cells by osterix promoter-driven Cre-recombinase, bone mineral density (BMD) was significantly reduced compared with floxed control littermates. Microcomputed tomography analysis also demonstrated a decrease in trabecular thickness and bone volume fraction in cKO mice at these ages. Dynamic bone histomorphometry suggested a mineralization defect in the cKO mice. However, by 9 months of age, the bone appeared to compensate for the lack of PRKD1, and BMD was not different. Taken together, these results suggest a potentially important role for PRKD1 in bone formation.
Collapse
Affiliation(s)
- Wendy B Bollag
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Physiology, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Medicine, Augusta University, 30912, United States; Department of Oral Biology, Augusta University, 30912, United States; Department of Cellular Biology and Anatomy, Augusta University, 30912, United States.
| | - Vivek Choudhary
- Charlie Norwood VA Medical Center, Augusta, GA 30904, United States; Department of Physiology, Augusta University, 30912, United States
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Ranya Elsayed
- Department of Oral Biology, Augusta University, 30912, United States
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Augusta University, 30912, United States
| | - Yun Su
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Lakiea J Bailey
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Xing-Ming Shi
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| | - Mohammed Elsalanty
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Oral Biology, Augusta University, 30912, United States
| | - Maribeth H Johnson
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States; Department of Biostatistics and Epidemiology, Augusta University, 30912, United States
| | - Meghan E McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Cellular Biology and Anatomy, Augusta University, 30912, United States
| | - Carlos M Isales
- Institute for Regenerative and Reparative Medicine, Augusta University, 30912, United States; Department of Orthopaedic Surgery, Augusta University, 30912, United States; Department of Neuroscience and Regenerative Medicine, Augusta University, 30912, United States
| |
Collapse
|
32
|
Feigenson M, Shull LC, Taylor EL, Camilleri ET, Riester SM, van Wijnen AJ, Bradley EW, Westendorf JJ. Histone Deacetylase 3 Deletion in Mesenchymal Progenitor Cells Hinders Long Bone Development. J Bone Miner Res 2017; 32:2453-2465. [PMID: 28782836 PMCID: PMC5732041 DOI: 10.1002/jbmr.3236] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/19/2017] [Accepted: 08/03/2017] [Indexed: 01/21/2023]
Abstract
Long bone formation is a complex process that requires precise transcriptional control of gene expression programs in mesenchymal progenitor cells. Histone deacetylases (Hdacs) coordinate chromatin structure and gene expression by enzymatically removing acetyl groups from histones and other proteins. Hdac inhibitors are used clinically to manage mood disorders, cancers, and other conditions but are teratogenic to the developing skeleton and increase fracture risk in adults. In this study, the functions of Hdac3, one of the enzymes blocked by current Hdac inhibitor therapies, in skeletal mesenchymal progenitor cells were determined. Homozygous deletion of Hdac3 in Prrx1-expressing cells prevented limb lengthening, altered pathways associated with endochondral and intramembranous bone development, caused perinatal lethality, and slowed chondrocyte and osteoblast differentiation in vitro. Transcriptomic analysis revealed that Hdac3 regulates vastly different pathways in mesenchymal cells expressing the Prxx1-Cre driver than those expressing the Col2-CreERT driver. Notably, Fgf21 was elevated in Hdac3-CKOPrrx1 limbs as well as in chondrogenic cells exposed to Hdac3 inhibitors. Elevated expression of Mmp3 and Mmp10 transcripts was also observed. In conclusion, Hdac3 regulates distinct pathways in mesenchymal cell populations and is required for mesenchymal progenitor cell differentiation and long bone development. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Marina Feigenson
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Lomeli Carpio Shull
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Earnest L Taylor
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | | | - Scott M Riester
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Andre J van Wijnen
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
33
|
El Refaey M, McGee-Lawrence ME, Fulzele S, Kennedy EJ, Bollag WB, Elsalanty M, Zhong Q, Ding KH, Bendzunas NG, Shi XM, Xu J, Hill WD, Johnson MH, Hunter M, Pierce JL, Yu K, Hamrick MW, Isales CM. Kynurenine, a Tryptophan Metabolite That Accumulates With Age, Induces Bone Loss. J Bone Miner Res 2017; 32:2182-2193. [PMID: 28727234 PMCID: PMC5685888 DOI: 10.1002/jbmr.3224] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Revised: 07/07/2017] [Accepted: 07/19/2017] [Indexed: 12/19/2022]
Abstract
Age-dependent bone loss occurs in humans and in several animal species, including rodents. The underlying causal mechanisms are probably multifactorial, although an age-associated increase in the generation of reactive oxygen species has been frequently implicated. We previously reported that aromatic amino acids function as antioxidants, are anabolic for bone, and that they may potentially play a protective role in an aging environment. We hypothesized that upon oxidation the aromatic amino acids would not only lose their anabolic effects but also potentially become a catabolic byproduct. When measured in vivo in C57BL/6 mice, the tryptophan oxidation product and kynurenine precursor, N-formylkynurenine (NFK), was found to increase with age. We tested the direct effects of feeding kynurenine (kyn) on bone mass and also tested the short-term effects of intraperitoneal kyn injection on bone turnover in CD-1 mice. μCT analyses showed kyn-induced bone loss. Levels of serum markers of osteoclastic activity (pyridinoline [PYD] and RANKL) increased significantly with kyn treatment. In addition, histological and histomorphometric studies showed an increase in osteoclastic activity in the kyn-treated groups in both dietary and injection-based studies. Further, kyn treatment significantly increased bone marrow adiposity, and BMSCs isolated from the kyn-injected mice exhibited decreased mRNA expression of Hdac3 and its cofactor NCoR1 and increased expression of lipid storage genes Cidec and Plin1. A similar pattern of gene expression is observed with aging. In summary, our data show that increasing kyn levels results in accelerated skeletal aging by impairing osteoblastic differentiation and increasing osteoclastic resorption. These data would suggest that kyn could play a role in age-induced bone loss. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Mona El Refaey
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - Meghan E. McGee-Lawrence
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Sadanand Fulzele
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
| | - Eileen J. Kennedy
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA, 30602
| | - Wendy B. Bollag
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
- Department of Physiology, Augusta University, Augusta, Georgia, 30912
- Department of Oral Biology, Augusta University, Augusta, Georgia, 30912
- Department of Charlie Norwood VA Medical Center, Augusta, Georgia, 30912
| | - Mohammed Elsalanty
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Oral Biology, Augusta University, Augusta, Georgia, 30912
| | - Qing Zhong
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - Ke-Hong Ding
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - Nathaniel G. Bendzunas
- Department of Pharmaceutical and Biomedical Sciences, University of Georgia College of Pharmacy, Athens, GA, 30602
| | - Xing-ming Shi
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
| | - Jianrui Xu
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
| | - William D. Hill
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
- Department of Charlie Norwood VA Medical Center, Augusta, Georgia, 30912
| | - Maribeth H. Johnson
- Department of Biostatistics and Epidemiology, Augusta University, Augusta, Georgia, 30912
| | - Monte Hunter
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
| | - Jessica L. Pierce
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Mark W. Hamrick
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| | - Carlos M. Isales
- Institute for Regenerative and Reparative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Neuroscience and Regenerative Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Orthopaedic Surgery, Augusta University, Augusta, Georgia, 30912
- Department of Medicine, Augusta University, Augusta, Georgia, 30912
- Department of Cellular Biology and Anatomy, Augusta University, Augusta, Georgia, 30912
| |
Collapse
|
34
|
Yu K, Doherty AH, Genik PC, Gookin SE, Roteliuk DM, Wojda SJ, Jiang ZS, McGee-Lawrence ME, Weil MM, Donahue SW. Mimicking the effects of spaceflight on bone: Combined effects of disuse and chronic low-dose rate radiation exposure on bone mass in mice. LIFE SCIENCES IN SPACE RESEARCH 2017; 15:62-68. [PMID: 29198315 DOI: 10.1016/j.lssr.2017.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 07/31/2017] [Accepted: 08/11/2017] [Indexed: 06/07/2023]
Abstract
During spaceflight, crewmembers are subjected to biomechanical and biological challenges including microgravity and radiation. In the skeleton, spaceflight leads to bone loss, increasing the risk of fracture. Studies utilizing hindlimb suspension (HLS) as a ground-based model of spaceflight often neglect the concomitant effects of radiation exposure, and even when radiation is accounted for, it is often delivered at a high-dose rate over a very short period of time, which does not faithfully mimic spaceflight conditions. This study was designed to investigate the skeletal effects of low-dose rate gamma irradiation (8.5 cGy gamma radiation per day for 20 days, amounting to a total dose of 1.7 Gy) when administered simultaneously to disuse from HLS. The goal was to determine whether continuous, low-dose rate radiation administered during disuse would exacerbate bone loss in a murine HLS model. Four groups of 16 week old female C57BL/6 mice were studied: weight bearing + no radiation (WB+NR), HLS + NR, WB + radiation exposure (WB+RAD), and HLS+RAD. Surprisingly, although HLS led to cortical and trabecular bone loss, concurrent radiation exposure did not exacerbate these effects. Our results raise the possibility that mechanical unloading has larger effects on the bone loss that occurs during spaceflight than low-dose rate radiation.
Collapse
Affiliation(s)
- Kanglun Yu
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China; Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Alison H Doherty
- Department of Medical Education, WWAMI Medical Education Program, University of Wyoming, Laramie, WY, USA
| | - Paula C Genik
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Sara E Gookin
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Danielle M Roteliuk
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Samantha J Wojda
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA
| | - Zhi-Sheng Jiang
- Institute of Cardiovascular Disease, Key Lab for Arteriosclerology of Hunan Province, University of South China, Hengyang, Hunan, China
| | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA, USA; Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, USA
| | - Michael M Weil
- Department of Environmental and Radiological Health Sciences, Colorado State University, Fort Collins, CO, USA
| | - Seth W Donahue
- Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, USA.
| |
Collapse
|
35
|
MiR-455-3p activates Nrf2/ARE signaling via HDAC2 and protects osteoblasts from oxidative stress. Int J Biol Macromol 2017; 107:2094-2101. [PMID: 29042277 DOI: 10.1016/j.ijbiomac.2017.10.080] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/13/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
BACKGROUND The important role of miR-455-3p in the pathogenesis of bone metabolism associated diseases is gradually emerging. This study aims to ascertain the involvement of miR-455-3p and its underlying mechanisms in osteoporosis. METHODS The osteoblast cell lines MC3T3-E1 was treated with ferric ammonium citrate (FAC) to mimic a pathological environment for osteoporosis. The cytotoxic effect of iron overload was assessed by proliferation, apoptosis and oxidative stress of osteoblasts using commercial kits. Molecular biological methods, including qRT-PCR analysis, cell transfection and luciferase reporter assays were used to explain the role of miR-455-3p and its potential mechanisms in osteoblast apoptosis. RESULTS FAC dramatically inhibited the proliferation of osteoblast cells MC3T3-E1 but increased the apoptosis. We also observed that FAC significantly down-regulated miR-455-3p in MC3T3-E1 cells but enhanced HDAC2 protein level. Moreover, miR-455-3p overexpression eliminated the effects of iron overload on osteoblast cell proliferation, apoptosis and oxidative stress. In addition, miR-455-3p regulated osteoblast cell proliferation, apoptosis and oxidative stress through regulating HDAC2-Nrf2/ARE signaling pathway. MiR-455-3p overexpression alleviated the oxidative stress injury in osteoporosis mice. CONCLUSION Our results demonstrated that miR-455-3p activated Nrf2/ARE signal pathway through suppressing Keap1 via negative regulating HDAC2 protein level, thereby suppressing oxidative stress and promoting osteoblasts growth.
Collapse
|
36
|
Abstract
PURPOSE OF REVIEW Bone remodeling is a diverse field of study with many direct clinical applications; past studies have implicated epigenetic alterations as key factors of both normal bone tissue development and function and diseases of pathologic bone remodeling. The purpose of this article is to review the most important recent advances that link epigenetic changes to the bone remodeling field. RECENT FINDINGS Epigenetics describes three major phenomena: DNA modification via methylation, histone side chain modifications, and short non-coding RNA sequences which work in concert to regulate gene transcription in a heritable fashion. Recent findings include the role of DNA methylation changes of Wnt, RANK/RANKL, and other key signaling pathways, epigenetic regulation of osteoblast and osteoclast differentiation, and others. Although much work has been done, much is still unknown. Future epigenome-wide studies should focus on extending the tissue coverage, integrating multiple epigenetic analyses with transcriptome data, and working to uncover epigenetic changes linked with early events in aberrant bone remodeling.
Collapse
Affiliation(s)
- Ali Husain
- Division of Rheumatology, Immunology, and Allergy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Matlock A Jeffries
- Division of Rheumatology, Immunology, and Allergy, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.
- Oklahoma Medical Research Foundation, Arthritis and Clinical Immunology Program, 825 NE 13th St., Laboratory MC400, Oklahoma City, OK, USA.
| |
Collapse
|
37
|
McGee-Lawrence ME, Pierce JL, Yu K, Culpepper NR, Bradley EW, Westendorf JJ. Loss of Hdac3 in osteoprogenitors increases bone expression of osteoprotegerin, improving systemic insulin sensitivity. J Cell Physiol 2017; 233:2671-2680. [PMID: 28840938 DOI: 10.1002/jcp.26148] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Accepted: 08/14/2017] [Indexed: 12/22/2022]
Abstract
Type 2 diabetes is an emerging global health epidemic. Foundations for new therapies are arising from understanding interactions between body systems. Bone-derived factors that reduce RANKL (receptor activator of NF-kappa B ligand) signaling in the liver may prevent insulin resistance and the onset of type 2 diabetes. Here we demonstrate that deletion of the epigenetic regulator, Hdac3, in Osx1-expressing osteoprogenitors prevents insulin resistance induced by high fat diet by increasing serum and skeletal gene expression levels of osteoprotegerin (Opg), a natural inhibitor of RANKL signaling. Removal of one Opg allele in mice lacking Hdac3 in Osx1+ osteoprogenitors increases the insulin resistance of the Hdac3-deficient mice on a high fat diet. Thus, Hdac3-depletion in osteoblasts increases expression of Opg, subsequently preserving insulin sensitivity. The Hdac inhibitor vorinostat also increased Opg transcription and histone acetylation of the Opg locus. These results define a new mechanism by which bone regulates systemic insulin sensitivity.
Collapse
Affiliation(s)
- Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia.,Department of Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jessica L Pierce
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Kanglun Yu
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Natasha R Culpepper
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia
| | | | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, Minnesota.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
38
|
Targeting cellular senescence prevents age-related bone loss in mice. Nat Med 2017; 23:1072-1079. [PMID: 28825716 PMCID: PMC5657592 DOI: 10.1038/nm.4385] [Citation(s) in RCA: 855] [Impact Index Per Article: 106.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 07/13/2017] [Indexed: 11/08/2022]
Abstract
Aging is associated with increased cellular senescence, which is hypothesized to drive the eventual development of multiple comorbidities. Here we investigate a role for senescent cells in age-related bone loss through multiple approaches. In particular, we used either genetic (i.e., the INK-ATTAC 'suicide' transgene encoding an inducible caspase 8 expressed specifically in senescent cells) or pharmacological (i.e., 'senolytic' compounds) means to eliminate senescent cells. We also inhibited the production of the proinflammatory secretome of senescent cells using a JAK inhibitor (JAKi). In aged (20- to 22-month-old) mice with established bone loss, activation of the INK-ATTAC caspase 8 in senescent cells or treatment with senolytics or the JAKi for 2-4 months resulted in higher bone mass and strength and better bone microarchitecture than in vehicle-treated mice. The beneficial effects of targeting senescent cells were due to lower bone resorption with either maintained (trabecular) or higher (cortical) bone formation as compared to vehicle-treated mice. In vitro studies demonstrated that senescent-cell conditioned medium impaired osteoblast mineralization and enhanced osteoclast-progenitor survival, leading to increased osteoclastogenesis. Collectively, these data establish a causal role for senescent cells in bone loss with aging, and demonstrate that targeting these cells has both anti-resorptive and anabolic effects on bone. Given that eliminating senescent cells and/or inhibiting their proinflammatory secretome also improves cardiovascular function, enhances insulin sensitivity, and reduces frailty, targeting this fundamental mechanism to prevent age-related bone loss suggests a novel treatment strategy not only for osteoporosis, but also for multiple age-related comorbidities.
Collapse
|
39
|
Huynh NCN, Everts V, Ampornaramveth RS. Histone deacetylases and their roles in mineralized tissue regeneration. Bone Rep 2017; 7:33-40. [PMID: 28856178 PMCID: PMC5565747 DOI: 10.1016/j.bonr.2017.08.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2017] [Revised: 04/19/2017] [Accepted: 08/09/2017] [Indexed: 01/18/2023] Open
Abstract
Histone acetylation is an important epigenetic mechanism that controls expression of certain genes. It includes non-sequence-based changes of chromosomal regional structure that can alter the expression of genes. Acetylation of histones is controlled by the activity of two groups of enzymes: the histone acetyltransferases (HATs) and histone deacetylases (HDACs). HDACs remove acetyl groups from the histone tail, which alters its charge and thus promotes compaction of DNA in the nucleosome. HDACs render the chromatin structure into a more compact form of heterochromatin, which makes the genes inaccessible for transcription. By altering the transcriptional activity of bone-associated genes, HDACs control both osteogenesis and osteoclastogenesis. This review presents an overview of the function of HDACs in the modulation of bone formation. Special attention is paid to the use of HDAC inhibitors in mineralized tissue regeneration from cells of dental origin. HDACs regulate the transcription activity of bone related genes. Inhibition of HDAC promotes osteogenic/odontogenic differentiation. HDAC inhibitors are applicable for mineral tissue regeneration therapy.
Collapse
Key Words
- ADSCs, adipose tissue-derived stem cells
- ALP, alkaline phosphatase
- BSP, bone sialoprotein
- Bone regeneration
- COL1, type I collagen
- DMP1, dentin matrix acidic phosphoprotein 1
- DPSCs, dental-derived stem cells
- DSPP, dentin sialophosphoprotein
- Dentin formation
- Epigenetic
- GSK-3, glycogen synthase kinase
- HAT, histone acetyltransferase
- HDAC, histone deacetylase
- Histone acetyltransferase
- Histone deacetylase
- MSCs, mesenchymal stem cells
- NaB, sodium butyrate
- OCN, osteocalcin
- OPN, osteopontin
- PCL/PEG, polycaprolactone/polyethylene glycol
- RUNX2, runt-related transcription factor 2
- SOST, sclerostin
- TGF-β/BMP, transforming growth factor-β/bone morphogenetic protein
- TSA, Trichostatin A
- VPA, valproic acid
- WNT/β-catenin, Wingless-int
- hPDLCs, human periodontal ligament cells
Collapse
Affiliation(s)
- Nam Cong-Nhat Huynh
- Department of Dental Basic Sciences, Faculty of Odonto-Stomatology, University of Medicine and Pharmacy at Ho Chi Minh City, Viet Nam
| | - Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Research Institute MOVE, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | | |
Collapse
|
40
|
Li J, Yang S, Li X, Liu D, Wang Z, Guo J, Tan N, Gao Z, Zhao X, Zhang J, Gou F, Yokota H, Zhang P. Role of endoplasmic reticulum stress in disuse osteoporosis. Bone 2017; 97:2-14. [PMID: 27989543 DOI: 10.1016/j.bone.2016.12.009] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2016] [Revised: 10/28/2016] [Accepted: 12/14/2016] [Indexed: 12/14/2022]
Abstract
Osteoporosis is a major skeletal disease with low bone mineral density, which leads to an increased risk of bone fracture. Salubrinal is a synthetic chemical that inhibits dephosphorylation of eukaryotic translation initiation factor 2 alpha (eIF2α) in response to endoplasmic reticulum (ER) stress. To understand possible linkage of osteoporosis to ER stress, we employed an unloading mouse model and examined the effects of salubrinal in the pathogenesis of disuse osteoporosis. The results presented several lines of evidence that osteoclastogenesis in the development of osteoporosis was associated with ER stress, and salubrinal suppressed unloading-induced bone loss. Compared to the age-matched control, unloaded mice reduced the trabecular bone area/total area (B.Ar/T.Ar) as well as the number of osteoblasts, and they increased the osteoclasts number on the trabecular bone surface in a time-dependent way. Unloading-induced disuse osteoporosis significantly increased the expression of Bip, p-eIF2α and ATF4 in short-term within 6h of tail suspension, but time-dependent decreased in HU2d to HU14d. Furthermore, a significant correlation of ER stress with the differentiation of osteoblasts and osteoclasts was observed. Administration of salubrinal suppressed the unloading-induced decrease in bone mineral density, B.Ar/T.Ar and mature osteoclast formation. Salubrinal also increased the colony-forming unit-fibroblasts and colony-forming unit-osteoblasts. It reduced the formation of mature osteoclasts, suppressed their migration and adhesion, and increased the expression of Bip, p-eIF2α and ATF4. Electron microscopy showed that rough endoplasmic reticulum expansion and a decreased number of ribosomes on ER membrane were observed in osteoblast of unloading mice, and the abnormal ER expansion was significantly improved by salubrinal treatment. A TUNEL assay together with CCAAT/enhancer binding protein homologous protein (CHOP) expression indicated that ER stress-induced osteoblast apoptosis was rescued by salubrinal. Collectively, the results support the notion that ER stress plays a key role in the pathogenesis of disuse osteoporosis, and salubrinal attenuates unloading-induced bone loss by altering proliferation and differentiation of osteoblasts and osteoclasts via eIF2α signaling.
Collapse
Affiliation(s)
- Jie Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300457, China
| | - Shuang Yang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300457, China
| | - Xinle Li
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300457, China; Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China
| | - Daquan Liu
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300457, China; Department of Pharmacology, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin 300100, China
| | - Zhaonan Wang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jialu Guo
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Nian Tan
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Zhe Gao
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Xiaoyu Zhao
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Jiuguo Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Fanglin Gou
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China
| | - Hiroki Yokota
- Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, IN 46202, USA
| | - Ping Zhang
- Department of Anatomy and Histology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin 300070, China; TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Tianjin 300457, China; Key Laboratory of Hormones and Development (Ministry of Health), Tianjin Key Laboratory of Metabolic Diseases, Tianjin Medical University, Tianjin 300070, China; Department of Biomedical Engineering, Indiana University-Purdue University Indianapolis, IN 46202, USA.
| |
Collapse
|
41
|
Cantley MD, Zannettino ACW, Bartold PM, Fairlie DP, Haynes DR. Histone deacetylases (HDAC) in physiological and pathological bone remodelling. Bone 2017; 95:162-174. [PMID: 27913271 DOI: 10.1016/j.bone.2016.11.028] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Revised: 10/31/2016] [Accepted: 11/28/2016] [Indexed: 11/21/2022]
Abstract
Histone deacetylases (HDACs)2 play important roles in the epigenetic regulation of gene expression in cells and are emerging therapeutic targets for treating a wide range of diseases. HDAC inhibitors (HDACi)3 that act on multiple HDAC enzymes have been used clinically to treat a number of solid and hematological malignancies. HDACi are also currently being studied for their efficacy in non-malignant diseases, including pathologic bone loss, but this has necessitated a better understanding of the roles of individual HDAC enzymes, particularly the eleven zinc-containing isozymes. Selective isozyme-specific inhibitors currently being developed against class I HDACs (1, 2, 3 and 8) and class II HDACs (4, 5, 6, 7, 9 and 10) will be valuable tools for elucidating the roles played by individual HDACs in different physiological and pathological settings. Isozyme-specific HDACi promise to have greater efficacy and reduced side effects, as required for treating chronic disease over extended periods of time. This article reviews the current understanding of roles for individual HDAC isozymes and effects of HDACi on bone cells, (osteoblasts, osteoclasts and osteocytes), in relation to bone remodelling in conditions characterised by pathological bone loss, including periodontitis, rheumatoid arthritis and myeloma bone disease.
Collapse
Affiliation(s)
- M D Cantley
- Discipline of Physiology, School of Medicine, University of Adelaide, SA 5005, Australia; Myeloma Research Laboratory, Cancer Theme, SAHMRI, Adelaide, SA 5000, Australia; Colgate Australian Clinical Dental Research Centre, Adelaide Dental School, University of Adelaide, SA 5005, Australia.
| | - A C W Zannettino
- Discipline of Physiology, School of Medicine, University of Adelaide, SA 5005, Australia; Myeloma Research Laboratory, Cancer Theme, SAHMRI, Adelaide, SA 5000, Australia
| | - P M Bartold
- Colgate Australian Clinical Dental Research Centre, Adelaide Dental School, University of Adelaide, SA 5005, Australia
| | - D P Fairlie
- Centre for Inflammation and Disease Research, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia; Australian Research Council Centre of Excellence in Advanced Molecular Imaging, Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - D R Haynes
- Discipline of Anatomy and Pathology, School of Medicine, University of Adelaide, SA 5005, Australia
| |
Collapse
|
42
|
Carpio LR, Bradley EW, Westendorf JJ. Histone deacetylase 3 suppresses Erk phosphorylation and matrix metalloproteinase (Mmp)-13 activity in chondrocytes. Connect Tissue Res 2017; 58:27-36. [PMID: 27662443 PMCID: PMC5609188 DOI: 10.1080/03008207.2016.1236088] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Histone deacetylase (Hdac3) inhibitors are emerging therapies for many diseases including cancers and neurological disorders; however, these drugs are teratogens to the developing skeleton. Hdac3 is essential for proper endochondral ossification as its deletion in chondrocytes increases cytokine signaling and the expression of matrix remodeling enzymes. Here we explored the mechanism by which Hdac3 controls matrix metalloproteinase (Mmp)-13 expression in chondrocytes. In Hdac3-depleted chondrocytes, extracellular signal-regulated kinase (Erk)1/2 as well as its downstream substrate, Runx2, were hyperphosphorylated as a result of decreased expression and activity of the Erk1/2 specific phosphatase, Dusp6. Erk1/2 kinase inhibitors and Dusp6 adenoviruses reduced Mmp13 expression and partially rescued matrix production in Hdac3-deficient chondrocytes. Postnatal chondrocyte-specific deletion of Hdac3 with an inducible Col2a1-Cre caused premature production of pErk1/2 and Mmp13 in the growth plate. Thus, Hdac3 controls the temporal and spatial expression of tissue-remodeling genes in chondrocytes to ensure proper endochondral ossification during development.
Collapse
Affiliation(s)
- Lomeli R. Carpio
- Mayo Graduate School, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | | | - Jennifer J. Westendorf
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
43
|
Carpio LR, Bradley EW, McGee-Lawrence ME, Weivoda MM, Poston DD, Dudakovic A, Xu M, Tchkonia T, Kirkland JL, van Wijnen AJ, Oursler MJ, Westendorf JJ. Histone deacetylase 3 supports endochondral bone formation by controlling cytokine signaling and matrix remodeling. Sci Signal 2016; 9:ra79. [PMID: 27507649 PMCID: PMC5409103 DOI: 10.1126/scisignal.aaf3273] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Histone deacetylase (HDAC) inhibitors are efficacious epigenetic-based therapies for some cancers and neurological disorders; however, each of these drugs inhibits multiple HDACs and has detrimental effects on the skeleton. To better understand how HDAC inhibitors affect endochondral bone formation, we conditionally deleted one of their targets, Hdac3, pre- and postnatally in type II collagen α1 (Col2α1)-expressing chondrocytes. Embryonic deletion was lethal, but postnatal deletion of Hdac3 delayed secondary ossification center formation, altered maturation of growth plate chondrocytes, and increased osteoclast activity in the primary spongiosa. HDAC3-deficient chondrocytes exhibited increased expression of cytokine and matrix-degrading genes (Il-6, Mmp3, Mmp13, and Saa3) and a reduced abundance of genes related to extracellular matrix production, bone development, and ossification (Acan, Col2a1, Ihh, and Col10a1). Histone acetylation increased at and near genes that had increased expression. The acetylation and activation of nuclear factor κB (NF-κB) were also increased in HDAC3-deficient chondrocytes. Increased cytokine signaling promoted autocrine activation of Janus kinase (JAK)-signal transducer and activator of transcription (STAT) and NF-κB pathways to suppress chondrocyte maturation, as well as paracrine activation of osteoclasts and bone resorption. Blockade of interleukin-6 (IL-6)-JAK-STAT signaling, NF-κB signaling, and bromodomain extraterminal proteins, which recognize acetylated lysines and promote transcriptional elongation, significantly reduced Il-6 and Mmp13 expression in HDAC3-deficient chondrocytes and secondary activation in osteoclasts. The JAK inhibitor ruxolitinib also reduced osteoclast activity in Hdac3 conditional knockout mice. Thus, HDAC3 controls the temporal and spatial expression of tissue-remodeling genes and inflammatory responses in chondrocytes to ensure proper endochondral ossification during development.
Collapse
Affiliation(s)
- Lomeli R Carpio
- Mayo Graduate School, Mayo Clinic, Rochester, MN 55905, USA. Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | | | - Meghan E McGee-Lawrence
- Department of Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA. Institute of Regenerative and Reparative Medicine, Augusta University, Augusta, GA 30912, USA
| | - Megan M Weivoda
- Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel D Poston
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Creighton University, Omaha, NE 68102, USA
| | - Amel Dudakovic
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Ming Xu
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Tamar Tchkonia
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - James L Kirkland
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN 55905, USA
| | - Andre J van Wijnen
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA
| | - Merry Jo Oursler
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Division of Endocrinology, Diabetes, Metabolism and Nutrition, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Jennifer J Westendorf
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA. Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
44
|
McGee-Lawrence ME, Carpio LR, Schulze RJ, Pierce JL, McNiven MA, Farr JN, Khosla S, Oursler MJ, Westendorf JJ. Hdac3 Deficiency Increases Marrow Adiposity and Induces Lipid Storage and Glucocorticoid Metabolism in Osteochondroprogenitor Cells. J Bone Miner Res 2016; 31. [PMID: 26211746 PMCID: PMC4758691 DOI: 10.1002/jbmr.2602] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Bone loss and increased marrow adiposity are hallmarks of aging skeletons. Conditional deletion of histone deacetylase 3 (Hdac3) in murine osteochondroprogenitor cells causes osteopenia and increases marrow adiposity, even in young animals, but the origins of the increased adiposity are unclear. To explore this, bone marrow stromal cells (BMSCs) from Hdac3-depleted and control mice were cultured in osteogenic medium. Hdac3-deficient cultures accumulated lipid droplets in greater abundance than control cultures and expressed high levels of genes related to lipid storage (Fsp27/Cidec, Plin1) and glucocorticoid metabolism (Hsd11b1) despite normal levels of Pparγ2. Approximately 5% of the lipid containing cells in the wild-type cultures expressed the master osteoblast transcription factor Runx2, but this population was threefold greater in the Hdac3-depleted cultures. Adenoviral expression of Hdac3 restored normal gene expression, indicating that Hdac3 controls glucocorticoid activation and lipid storage within osteoblast lineage cells. HDAC3 expression was reduced in bone cells from postmenopausal as compared to young women, and in osteoblasts from aged as compared to younger mice. Moreover, phosphorylation of S424 in Hdac3, a posttranslational mark necessary for deacetylase activity, was suppressed in osseous cells from old mice. Thus, concurrent declines in transcription and phosphorylation combine to suppress Hdac3 activity in aging bone, and reduced Hdac3 activity in osteochondroprogenitor cells contributes to increased marrow adiposity associated with aging. © 2015 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Meghan E McGee-Lawrence
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Lomeli R Carpio
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| | - Ryan J Schulze
- Department of Medicine, Division of Gastroenterology and Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Jessica L Pierce
- Department of Cellular Biology and Anatomy, Georgia Regents University, Augusta, GA, USA
| | - Mark A McNiven
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Department of Medicine, Division of Gastroenterology and Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Joshua N Farr
- Department of Medicine, Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Sundeep Khosla
- Department of Medicine, Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Merry Jo Oursler
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA.,Department of Medicine, Division of Endocrinology, Mayo Clinic, Rochester, MN, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
45
|
Gordon JAR, Stein JL, Westendorf JJ, van Wijnen AJ. Chromatin modifiers and histone modifications in bone formation, regeneration, and therapeutic intervention for bone-related disease. Bone 2015; 81:739-745. [PMID: 25836763 PMCID: PMC4591092 DOI: 10.1016/j.bone.2015.03.011] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/13/2015] [Indexed: 02/07/2023]
Abstract
Post-translational modifications of chromatin such as DNA methylation and different types of histone acetylation, methylation and phosphorylation are well-appreciated epigenetic mechanisms that confer information to progeny cells during lineage commitment. These distinct epigenetic modifications have defined roles in bone, development, tissue regeneration, cell commitment and differentiation, as well as disease etiologies. In this review, we discuss the role of these chromatin modifications and the enzymes regulating these marks (methyltransferases, demethylases, acetyltransferases, and deacetylases) in progenitor cells, osteoblasts and bone-related cells. In addition, the clinical relevance of deregulated histone modifications and enzymes as well as current and potential therapeutic interventions targeting chromatin modifiers are addressed.
Collapse
Affiliation(s)
| | - Janet L Stein
- Department of Biochemistry, University of Vermont, Burlington, VT, USA.
| | | | | |
Collapse
|
46
|
Bradley EW, Carpio LR, van Wijnen AJ, McGee-Lawrence ME, Westendorf JJ. Histone Deacetylases in Bone Development and Skeletal Disorders. Physiol Rev 2015; 95:1359-81. [PMID: 26378079 PMCID: PMC4600951 DOI: 10.1152/physrev.00004.2015] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Histone deacetylases (Hdacs) are conserved enzymes that remove acetyl groups from lysine side chains in histones and other proteins. Eleven of the 18 Hdacs encoded by the human and mouse genomes depend on Zn(2+) for enzymatic activity, while the other 7, the sirtuins (Sirts), require NAD2(+). Collectively, Hdacs and Sirts regulate numerous cellular and mitochondrial processes including gene transcription, DNA repair, protein stability, cytoskeletal dynamics, and signaling pathways to affect both development and aging. Of clinical relevance, Hdacs inhibitors are United States Food and Drug Administration-approved cancer therapeutics and are candidate therapies for other common diseases including arthritis, diabetes, epilepsy, heart disease, HIV infection, neurodegeneration, and numerous aging-related disorders. Hdacs and Sirts influence skeletal development, maintenance of mineral density and bone strength by affecting intramembranous and endochondral ossification, as well as bone resorption. With few exceptions, inhibition of Hdac or Sirt activity though either loss-of-function mutations or prolonged chemical inhibition has negative and/or toxic effects on skeletal development and bone mineral density. Specifically, Hdac/Sirt suppression causes abnormalities in physiological development such as craniofacial dimorphisms, short stature, and bone fragility that are associated with several human syndromes or diseases. In contrast, activation of Sirts may protect the skeleton from aging and immobilization-related bone loss. This knowledge may prolong healthspan and prevent adverse events caused by epigenetic therapies that are entering the clinical realm at an unprecedented rate. In this review, we summarize the general properties of Hdacs/Sirts and the research that has revealed their essential functions in bone forming cells (e.g., osteoblasts and chondrocytes) and bone resorbing osteoclasts. Finally, we offer predictions on future research in this area and the utility of this knowledge for orthopedic applications and bone tissue engineering.
Collapse
Affiliation(s)
- Elizabeth W Bradley
- Mayo Clinic, Departments of Orthopedic Surgery and of Biochemistry and Molecular Biology, and Mayo Graduate School, Rochester, Minnesota; and Georgia Regents University, Department of Cellular Biology and Anatomy, Augusta, Georgia
| | - Lomeli R Carpio
- Mayo Clinic, Departments of Orthopedic Surgery and of Biochemistry and Molecular Biology, and Mayo Graduate School, Rochester, Minnesota; and Georgia Regents University, Department of Cellular Biology and Anatomy, Augusta, Georgia
| | - Andre J van Wijnen
- Mayo Clinic, Departments of Orthopedic Surgery and of Biochemistry and Molecular Biology, and Mayo Graduate School, Rochester, Minnesota; and Georgia Regents University, Department of Cellular Biology and Anatomy, Augusta, Georgia
| | - Meghan E McGee-Lawrence
- Mayo Clinic, Departments of Orthopedic Surgery and of Biochemistry and Molecular Biology, and Mayo Graduate School, Rochester, Minnesota; and Georgia Regents University, Department of Cellular Biology and Anatomy, Augusta, Georgia
| | - Jennifer J Westendorf
- Mayo Clinic, Departments of Orthopedic Surgery and of Biochemistry and Molecular Biology, and Mayo Graduate School, Rochester, Minnesota; and Georgia Regents University, Department of Cellular Biology and Anatomy, Augusta, Georgia
| |
Collapse
|
47
|
Ryan ZC, Craig TA, Filoteo AG, Westendorf JJ, Cartwright EJ, Neyses L, Strehler EE, Kumar R. Deletion of the intestinal plasma membrane calcium pump, isoform 1, Atp2b1, in mice is associated with decreased bone mineral density and impaired responsiveness to 1, 25-dihydroxyvitamin D3. Biochem Biophys Res Commun 2015; 467:152-6. [PMID: 26392310 DOI: 10.1016/j.bbrc.2015.09.087] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 09/16/2015] [Indexed: 01/01/2023]
Abstract
The physiological importance of the intestinal plasma membrane calcium pump, isoform 1, (Pmca1, Atp2b1), in calcium absorption and homeostasis has not been previously demonstrated in vivo. Since global germ-line deletion of the Pmca1 in mice is associated with embryonic lethality, we selectively deleted the Pmca1 in intestinal absorptive cells. Mice with loxP sites flanking exon 2 of the Pmca1 gene (Pmca1(fl/fl)) were crossed with mice expressing Cre recombinase in the intestine under control of the villin promoter to give mice in which the Pmca1 had been deleted in the intestine (Pmca1(EKO) mice). Pmca1(EKO) mice were born at a reduced frequency and were small at the time of birth when compared to wild-type (Wt) littermates. At two months of age, Pmca1(EKO) mice fed a 0.81% calcium, 0.34% phosphorus, normal vitamin D diet had reduced whole body bone mineral density (P < 0.037), and reduced femoral bone mineral density (P < 0.015). There was a trend towards lower serum calcium and higher serum parathyroid hormone (PTH) and 1α,25-dihydroxyvitamin D3 (1α,25(OH)2D3) concentrations in Pmca1(EKO) mice compared to Wt mice but the changes were not statistically significant. The urinary phosphorus/creatinine ratio was increased in Pmca1(EKO) mice (P < 0.004). Following the administration of 200 ng of 1α,25(OH)2D3 intraperitoneally to Wt mice, active intestinal calcium transport increased ∼2-fold, whereas Pmca1(EKO) mice administered an equal amount of 1α,25(OH)2D3 failed to show an increase in active calcium transport. Deletion of the Pmca1 in the intestine is associated with reduced growth and bone mineralization, and a failure to up-regulate calcium absorption in response to 1α,25(OH)2D3.
Collapse
Affiliation(s)
- Zachary C Ryan
- Nephrology Research, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Theodore A Craig
- Nephrology Research, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Adelaida G Filoteo
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA; Department of Orthopedic Surgery, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA
| | | | - Ludwig Neyses
- Institute of Cardiovascular Sciences, University of Manchester, Manchester, M13 9PT, UK; University of Luxembourg, L-4365, Esch-sur-Alzette, Luxembourg
| | - Emanuel E Strehler
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| | - Rajiv Kumar
- Nephrology Research, Department of Internal Medicine, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA; Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN, 55905, USA.
| |
Collapse
|
48
|
Silencing of RUNX2 enhances gemcitabine sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells through the stimulation of TAp63-mediated cell death. Cell Death Discov 2015; 1:15010. [PMID: 27551445 PMCID: PMC4981025 DOI: 10.1038/cddiscovery.2015.10] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 06/16/2015] [Indexed: 12/19/2022] Open
Abstract
Runt-related transcription factor 2 (RUNX2) has been considered to be one of master regulators for osteoblast differentiation and bone formation. Recently, we have described that RUNX2 attenuates p53/TAp73-dependent cell death of human osteosarcoma U2OS cells bearing wild-type p53 in response to adriamycin. In this study, we have asked whether RUNX2 silencing could enhance gemcitabine (GEM) sensitivity of p53-deficient human pancreatic cancer AsPC-1 cells. Under our experimental conditions, GEM treatment increased the expression level of p53 family TAp63, whereas RUNX2 was reduced following GEM exposure, indicating that there exists an inverse relationship between the expression level of TAp63 and RUNX2 following GEM exposure. To assess whether TAp63 could be involved in the regulation of GEM sensitivity of AsPC-1 cells, small interfering RNA-mediated knockdown of TAp63 was performed. As expected, silencing of TAp63 significantly prohibited GEM-dependent cell death as compared with GEM-treated non-silencing cells. As TAp63 was negatively regulated by RUNX2, we sought to examine whether RUNX2 knockdown could enhance the sensitivity to GEM. Expression analysis demonstrated that depletion of RUNX2 apparently stimulates the expression of TAp63, as well as proteolytic cleavage of poly ADP ribose polymerase (PARP) after GEM exposure, and further augmented GEM-mediated induction of p53/TAp63-target genes, such as p21WAF1, PUMA and NOXA, relative to GEM-treated control-transfected cells, implying that RUNX2 has a critical role in the regulation of GEM resistance through the downregulation of TAp63. Notably, ablation of TAp63 gave a decrease in number of γH2AX-positive cells in response to GEM relative to control-transfected cells following GEM exposure. Consistently, GEM-dependent phosphorylation of ataxia telangiectasia-mutated protein was remarkably impaired in TAp63 knockdown cells. Collectively, our present findings strongly suggest that RUNX2-mediated repression of TAp63 contributes at least in part to GEM resistance of AsPC-1 cells, and thus silencing of RUNX2 may be a novel strategy to enhance the efficacy of GEM in p53-deficient pancreatic cancer cells.
Collapse
|
49
|
The effect of laser therapy on the expression of osteocalcin and osteopontin after tooth extraction in rats treated with zoledronate and dexamethasone. Support Care Cancer 2015; 24:807-813. [PMID: 26190360 DOI: 10.1007/s00520-015-2847-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/06/2015] [Indexed: 12/20/2022]
Abstract
PURPOSE Laser therapy has been used for the prevention and management of medication-related ostenecrosis of the jaw (MRONJ). The aim of this paper was to investigate the action of laser therapy on extraction socket healing in rats in conditions at risk for MRONJ, evaluating the expression of markers of bone metabolism. METHODS Thirty male Sprague-Dawley rats were divided in four groups: control group (C, n = 5), laser group (L, n = 5), treatment group (T, n = 10), and treatment plus laser group (T + L, n = 10). Rats of group T and T + L received zoledronate 0.1 mg/kg and dexamethasone 1 mg/kg every 2 days for 10 weeks. Rats of group C and L were infused with vehicle. After 9 weeks, the left maxillary molars were extracted in all rats. Rats of groups L and T + L received laser therapy (Nd:YAG, 1064 nm, 1.25 W, 15 Hz, 5 min, 14.37 J/cm(2)) in the socket area at days 0, 2, 4, and 6 after surgery. Western blot analysis was performed to evaluate the alveolar expression of osteopontin (OPN) and osteocalcin (OCN) 8 days after extraction. RESULTS Rats of groups L and T + L showed a significant higher expression of OCN compared to rats of groups C and T (+348 and +400 %, respectively; P = 0.013 and P = 0.002, respectively). The expression of OPN did not show significant differences among the different groups. CONCLUSIONS Our findings suggest that laser irradiation after tooth extraction can promote osteoblast differentiation, as demonstrated by the higher expression of OCN. Thus, laser irradiation could be considered a way to improve socket healing in conditions at risk for MRONJ development.
Collapse
|
50
|
McGee-Lawrence ME, White TA, LeBrasseur NK, Westendorf JJ. Conditional deletion of Hdac3 in osteoprogenitor cells attenuates diet-induced systemic metabolic dysfunction. Mol Cell Endocrinol 2015; 410:42-51. [PMID: 25666992 PMCID: PMC4444388 DOI: 10.1016/j.mce.2015.02.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2014] [Revised: 02/03/2015] [Accepted: 02/03/2015] [Indexed: 01/30/2023]
Abstract
Obesity is a major health epidemic in the United States and a leading cause of preventable diseases including type 2 diabetes. A growing body of evidence indicates that the skeleton influences whole body metabolism and suggests a new avenue for developing novel therapeutic agents, but the underlying mechanisms are not well understood. Here, it is demonstrated that conditional deletion of an epigenetic regulator, Hdac3, in osteoblast progenitor cells abrogates high fat diet-induced insulin resistance and hepatic steatosis. These Hdac3-deficient mice have reduced bone formation and lower circulating levels of total and undercarboxylated osteocalcin, coupled with decreased bone resorption activity. They also maintain lower body fat and fasting glucose levels on normal and high fat chow diets. The mechanisms by which Hdac3 controls systemic energy homeostasis from within osteoblasts have not yet been fully realized, but the current study suggests that it does not involve elevated levels of circulating osteocalcin. Thus, Hdac3 is a new player in the emerging paradigm that the skeleton influences systemic energy metabolism.
Collapse
|