1
|
Reversing the imbalance in bone homeostasis via sustained release of SIRT-1 agonist to promote bone healing under osteoporotic condition. Bioact Mater 2023; 19:429-443. [PMID: 35574058 PMCID: PMC9079176 DOI: 10.1016/j.bioactmat.2022.04.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/14/2022] [Accepted: 04/15/2022] [Indexed: 12/12/2022] Open
Abstract
The imbalance of bone homeostasis is the root cause of osteoporosis. However current therapeutic approaches mainly focus on either anabolic or catabolic pathways, which often fail to turn the imbalanced bone metabolism around. Herein we reported that a SIRT-1 agonist mediated molecular therapeutic strategy to reverse the imbalance in bone homeostasis by simultaneously regulating osteogenesis and osteoclastogenesis via locally sustained release of SRT2104 from mineral coated acellular matrix microparticles. Immobilization of SRT2104 on mineral coating (MAM/SRT) harnessing their electrostatic interactions resulted in sustained release of SIRT-1 agonist for over 30 days. MAM/SRT not only enhanced osteogenic differentiation and mineralization, but also attenuated the formation and function of excessive osteoclasts via integrating multiple vital upstream signals (β-catenin, FoxOs, Runx2, NFATc1, etc.) in vitro. Osteoporosis animal model also validated that it accelerated osteoporotic bone healing and improved osseointegration of the surrounding bone. Overall, our work proposes a promising strategy to treat osteoporotic bone defects by reversing the imbalance in bone homeostasis using designated small molecule drug delivery systems. A mineral coated acellular matrix microcarriers sustainably release SIRT2104 more than 30 days. This drug delivery system regulates osteogenesis and osteoclastogenesis. It can accelerate osteoporotic bone healing by reversing the imbalance in bone homeostasis.
Collapse
|
2
|
Filipović M, Flegar D, Šućur A, Šisl D, Kavazović I, Antica M, Kelava T, Kovačić N, Grčević D. Inhibition of Notch Signaling Stimulates Osteoclastogenesis From the Common Trilineage Progenitor Under Inflammatory Conditions. Front Immunol 2022; 13:902947. [PMID: 35865541 PMCID: PMC9294223 DOI: 10.3389/fimmu.2022.902947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 05/25/2022] [Indexed: 11/13/2022] Open
Abstract
Osteoclasts, macrophages and dendritic cells (DCs) can be derived from a common trilineage myeloid progenitor of hematopoietic origin. Progenitor commitment is susceptible to regulation through Notch signaling. Our aim was to determine the effects of Notch modulation on trilineage progenitor commitment and functional properties of differentiated cells under inflammatory conditions. We used the conditional inducible CX3CR1CreERT2 mouse strain to achieve overexpression of the Notch 1 intracellular domain (NICD1) or to inhibit Notch signaling via deletion of the transcription factor RBP-J in a bone marrow population, used as a source of the trilineage progenitor (CD45+Ly6G−CD3−B220−NK1.1−CD11b–/loCD115+). Cre-recombinase, under the control of the CX3CR1 promoter, expressed in the monocyte/macrophage lineage, was induced in vitro by 4-hydroxytamoxifen. Differentiation of osteoclasts was induced by M-CSF/RANKL; macrophages by M-CSF; DCs by IL-4/GM-CSF, and inflammation by LPS. Functionally, DCs were tested for the ability to process and present antigen, macrophages to phagocytose E. coli particles, and osteoclasts to resorb bone and express tartrate-resistant acid phosphatase (TRAP). We found that Notch 1 signal activation suppressed osteoclast formation, whereas disruption of the Notch canonical pathway enhanced osteoclastogenesis, resulting in a higher number and size of osteoclasts. RANK protein and Ctsk gene expression were upregulated in osteoclastogenic cultures from RBP-J+ mice, with the opposing results in NICD1+ mice. Notch modulation did not affect the number of in vitro differentiated macrophages and DCs. However, RBP-J deletion stimulated Il12b and Cd86 expression in macrophages and DCs, respectively. Functional assays under inflammatory conditions confirmed that Notch silencing amplifies TRAP expression by osteoclasts, whereas the enhanced phagocytosis by macrophages was observed in both NICD1+ and RBP-J+ strains. Finally, antigen presentation by LPS-stimulated DCs was significantly downregulated with NICD1 overexpression. This experimental setting allowed us to define a cell-autonomous response to Notch signaling at the trilineage progenitor stage. Although Notch signaling modulation affected the activity of all three lineages, the major effect was observed in osteoclasts, resulting in enhanced differentiation and function with inhibition of canonical Notch signaling. Our results indicate that Notch signaling participates as the negative regulator of osteoclast activity during inflammation, which may be relevant in immune and bone diseases.
Collapse
Affiliation(s)
- Maša Filipović
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Darja Flegar
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Alan Šućur
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Dino Šisl
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Inga Kavazović
- Department of Histology and Embryology, Faculty of Medicine, University of Rijeka, Rijeka, Croatia
| | | | - Tomislav Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Nataša Kovačić
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia
- Laboratory for Molecular Immunology, Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
- *Correspondence: Danka Grčević,
| |
Collapse
|
3
|
Wang Y, Li X, Liu Y, Wang J, Huang X. Effect of MC3T3 cell density on osteoclastic differentiation of mouse bone marrow cells. Tissue Cell 2022; 75:101724. [DOI: 10.1016/j.tice.2021.101724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 11/26/2021] [Accepted: 12/27/2021] [Indexed: 10/19/2022]
|
4
|
Root SH, Aguila HL. Novel population of human monocyte and osteoclast progenitors from pluripotent stem cells and peripheral blood. Blood Adv 2021; 5:4435-4446. [PMID: 34581760 PMCID: PMC8579260 DOI: 10.1182/bloodadvances.2021004552] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 07/09/2021] [Indexed: 12/21/2022] Open
Abstract
Osteoclasts are multinuclear cells of monocytic lineage, with the ability to resorb bone. Studies in mouse have identified bone marrow clonal progenitors able to generate mature osteoclast cells (OCs) in vitro and in vivo. These osteoclast progenitors (OCPs) can also generate macrophages and dendritic cells. Interestingly, cells with equivalent potential can be detected in periphery. In humans, cells with OCP activity have been identified in bone marrow and periphery; however, their characterization has not been as extensive. We have developed reproducible methods to derive, from human pluripotent stem cells, a population containing monocyte progenitors able to generate functional OCs. Within this population, we have identified cells with monocyte and osteoclast progenitor activity based on CD11b and CD14 expression. A population double positive for CD11b and CD14 contains cells with expected osteoclastic potential. However, the double negative (DN) population, containing most of the hematopoietic progenitor activity, also presents a very high osteoclastic potential. These progenitor cells can also be differentiated to macrophage and dendritic cells. Further dissection within the DN population identified cells bearing the phenotype CD15-CD115+ as the population with highest monocytic progenitor and osteoclastic potential. When similar methodology was used to identify OCPs from human peripheral blood, we confirmed a published OCP population with the phenotype CD11b+CD14+. In addition, we identified a second population (CD14-CD11bloCD115+) with high monocytic progenitor activity that was also able to form osteoclast like cells, similar to the 2 populations identified from pluripotent stem cells.
Collapse
Affiliation(s)
- Sierra H. Root
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT
| | - Héctor L. Aguila
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT
| |
Collapse
|
5
|
Sun P, Wang M, Yin GY. Endogenous parathyroid hormone (PTH) signals through osteoblasts via RANKL during fracture healing to affect osteoclasts. Biochem Biophys Res Commun 2020; 525:850-856. [PMID: 32169280 DOI: 10.1016/j.bbrc.2020.02.177] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 02/29/2020] [Indexed: 11/28/2022]
Abstract
AIM To investigate the effect of endogenous PTH deficiency on osteoclasts during fracture healing and its mechanism. METHODS A femoral fracture model was used to determine the role of endogenous PTH in fracture healing. Immunohistochemistry, qPCR, and Western blot were used to determine the potential functions and mechanisms of endogenous PTH. RESULT In this study, we found that expression of RANKL and CK was lower in PTH knockout (KO) mice than in wild type (WT) mice. In vitro culture of osteoclasts showed that under the same stimulation, there was no statistical difference in the number of osteoclasts and the area of bone resorption areas in PTH WT mice and PTH KO mice. We found that a high concentration of RANKL could promote the number and activity of osteoclasts. Upon induction of osteoblasts in vitro, those from the PTH WT group expressed higher RANKL protein and mRNA than those from the PTH KO group. Lastly, we confirmed that the PI3K/AKT/STAT5 pathway promotes RANKL increase from osteoblasts. CONCLUSION During fracture healing, endogenous PTH deficiency can affect osteoclast activity by reducing RANKL expression in osteoblasts.
Collapse
Affiliation(s)
- Peng Sun
- Department of Orthopedics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, 223300, China
| | - Ming Wang
- Department of Plastic and Burn Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China
| | - Guo-Yong Yin
- Department of Orthopedics, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, 210029, China.
| |
Collapse
|
6
|
Nelson AJ, Roy SK, Warren K, Janike K, Thiele GM, Mikuls TR, Romberger DJ, Wang D, Swanson B, Poole JA. Sex differences impact the lung-bone inflammatory response to repetitive inhalant lipopolysaccharide exposures in mice. J Immunotoxicol 2018; 15:73-81. [PMID: 29648480 PMCID: PMC6122601 DOI: 10.1080/1547691x.2018.1460425] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Revised: 03/21/2018] [Accepted: 03/29/2018] [Indexed: 12/11/2022] Open
Abstract
Skeletal health consequences associated with inflammatory diseases of the airways significantly contribute to morbidity. Sex differences have been described independently for lung and bone diseases. Repetitive inhalant exposure to lipopolysaccharide (LPS) induces bone loss and deterioration in male mice, but comparison effects in females are unknown. Using an intranasal inhalation exposure model, 8-week-old C57BL/6 male and female mice were treated daily with LPS (100 ng) or saline for 3 weeks. Bronchoalveolar lavage fluids, lung tissues, tibias, bone marrow cells, and blood were collected. LPS-induced airway neutrophil influx, interleukin (IL)-6 and neutrophil chemoattractant levels, and bronchiolar inflammation were exaggerated in male animals as compared to female mice. Trabecular bone micro-CT imaging and analysis of the proximal tibia were conducted. Inhalant LPS exposures lead to deterioration of bone quality only in male mice (not females) marked by decreased bone mineral density, bone volume/tissue volume ratio, trabecular thickness and number, and increased bone surface-to-bone volume ratio. Serum pentraxin-2 levels were modulated by sex differences and LPS exposure. In proof-of-concept studies, ovarectomized female mice demonstrated LPS-induced bone deterioration, and estradiol supplementation of ovarectomized female mice and control male mice protected against LPS-induced bone deterioration findings. Collectively, sex-specific differences exist in LPS-induced airway inflammatory consequences with significant differences found in bone quantity and quality parameters. Male mice demonstrated susceptibility to bone loss and female animals were protected, which was modulated by estrogen. Therefore, sex differences influence the biologic response in the lung-bone inflammatory axis in response to inhalant LPS exposures.
Collapse
Affiliation(s)
- Amy J. Nelson
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center
| | - Shyamal K. Roy
- Department of Obstetrics and Gynecology, University of Nebraska Medical Center
| | - Kristi Warren
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center
| | - Katherine Janike
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center
- Department of Internal Medicine, Rheumatology Division, University of Nebraska Medical Center
| | - Geoffrey M. Thiele
- Veterans Affairs Nebraska-Western Iowa Health Care System
- Department of Internal Medicine, Rheumatology Division, University of Nebraska Medical Center
| | - Ted R. Mikuls
- Veterans Affairs Nebraska-Western Iowa Health Care System
- Department of Internal Medicine, Rheumatology Division, University of Nebraska Medical Center
| | - Debra J. Romberger
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center
- Veterans Affairs Nebraska-Western Iowa Health Care System
| | - Dong Wang
- Department of Internal Medicine, Pharmaceutical Sciences, University of Nebraska Medical Center
| | - Benjamin Swanson
- Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE
| | - Jill A. Poole
- Department of Internal Medicine, Pulmonary, Critical Care, Sleep & Allergy Division, University of Nebraska Medical Center
| |
Collapse
|
7
|
Grčević D, Sironi M, Valentino S, Deban L, Cvija H, Inforzato A, Kovačić N, Katavić V, Kelava T, Kalajzić I, Mantovani A, Bottazzi B. The Long Pentraxin 3 Plays a Role in Bone Turnover and Repair. Front Immunol 2018; 9:417. [PMID: 29556234 PMCID: PMC5845433 DOI: 10.3389/fimmu.2018.00417] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 02/15/2018] [Indexed: 01/04/2023] Open
Abstract
Pentraxin 3 (PTX3) is an inflammatory mediator acting as a fluid-phase pattern recognition molecule and playing an essential role in innate immunity and matrix remodeling. Inflammatory mediators also contribute to skeletal homeostasis, operating at multiple levels in physiological and pathological conditions. This study was designed to investigate the role of PTX3 in physiological skeletal remodeling and bone healing. Micro-computed tomography (μCT) and bone histomorphometry of distal femur showed that PTX3 gene-targeted female and male mice (ptx3−/−) had lower trabecular bone volume than their wild-type (ptx3+/+) littermates (BV/TV by μCT: 3.50 ± 1.31 vs 6.09 ± 1.17 for females, p < 0.0001; BV/TV 9.06 ± 1.89 vs 10.47 ± 1.97 for males, p = 0.0435). In addition, μCT revealed lower trabecular bone volume in second lumbar vertebra of ptx3−/− mice. PTX3 was increasingly expressed during osteoblast maturation in vitro and was able to reverse the negative effect of fibroblast growth factor 2 (FGF2) on osteoblast differentiation. This effect was specific for the N-terminal domain of PTX3 that contains the FGF2-binding site. By using the closed transversal tibial fracture model, we found that ptx3−/− female mice formed significantly less mineralized callus during the anabolic phase following fracture injury compared to ptx3+/+ mice (BV/TV 17.05 ± 4.59 vs 20.47 ± 3.32, p = 0.0195). Non-hematopoietic periosteal cells highly upregulated PTX3 expression during the initial phase of fracture healing, particularly CD51+ and αSma+ osteoprogenitor subsets, and callus tissue exhibited concomitant expression of PTX3 and FGF2 around the fracture site. Thus, PTX3 supports maintenance of the bone mass possibly by inhibiting FGF2 and its negative impact on bone formation. Moreover, PTX3 enables timely occurring sequence of callus mineralization after bone fracture injury. These results indicate that PTX3 plays an important role in bone homeostasis and in proper matrix mineralization during fracture repair, a reflection of the function of this molecule in tissue homeostasis and repair.
Collapse
Affiliation(s)
- Danka Grčević
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Marina Sironi
- Humanitas Clinical and Research Center, Milan, Italy
| | | | - Livija Deban
- Humanitas Clinical and Research Center, Milan, Italy.,Oxford BioTherapeutics Ltd., Abingdon, United Kingdom
| | - Hrvoje Cvija
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Antonio Inforzato
- Humanitas Clinical and Research Center, Milan, Italy.,Department of Medical Biotechnologies and Translational Medicine, University of Milan, Milan, Italy
| | - Nataša Kovačić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Vedran Katavić
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia.,Department of Anatomy, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Tomislav Kelava
- Department of Physiology and Immunology, University of Zagreb School of Medicine, Zagreb, Croatia.,Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Ivo Kalajzić
- Department of Reconstructive Sciences, School of Dental Medicine, UConn Health, Farmingam, CT, United States
| | - Alberto Mantovani
- Humanitas Clinical and Research Center, Milan, Italy.,Humanitas University, Milan, Italy.,The William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
| | | |
Collapse
|
8
|
Teti A, Econs MJ. Osteopetroses, emphasizing potential approaches to treatment. Bone 2017; 102:50-59. [PMID: 28167345 DOI: 10.1016/j.bone.2017.02.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 02/02/2017] [Accepted: 02/02/2017] [Indexed: 12/22/2022]
Abstract
Osteopetroses are a heterogeneous group of rare genetic bone diseases sharing the common hallmarks of reduced osteoclast activity, increased bone mass and high bone fragility. Osteoclasts are bone resorbing cells that contribute to bone growth and renewal through the erosion of the mineralized matrix. Alongside the bone forming activity by osteoblasts, osteoclasts allow the skeleton to grow harmonically and maintain a healthy balance between bone resorption and formation. Osteoclast impairment in osteopetroses prevents bone renewal and deteriorates bone quality, causing atraumatic fractures. Osteopetroses vary in severity and are caused by mutations in a variety of genes involved in bone resorption or in osteoclastogenesis. Frequent signs and symptoms include osteosclerosis, deformity, dwarfism and narrowing of the bony canals, including the nerve foramina, leading to hematological and neural failures. The disease is autosomal, with only one extremely rare form associated so far to the X-chromosome, and can have either recessive or dominant inheritance. Recessive ostepetroses are generally lethal in infancy or childhood, with a few milder forms clinically denominated intermediate osteopetroses. Dominant osteopetrosis is so far associated only with mutations in the CLCN7 gene and, although described as a benign form, it can be severely debilitating, although not at the same level as recessive forms, and can rarely result in reduced life expectancy. Severe osteopetroses due to osteoclast autonomous defects can be treated by Hematopoietic Stem Cell Transplant (HSCT), but those due to deficiency of the pro-osteoclastogenic cytokine, RANKL, are not suitable for this procedure. Likewise, it is unclear as to whether HSCT, which has high intrinsic risks, results in clinical improvement in autosomal dominant osteopetrosis. Therefore, there is an unmet medical need to identify new therapies and studies are currently in progress to test gene and cell therapies, small interfering RNA approach and novel pharmacologic treatments.
Collapse
Affiliation(s)
- Anna Teti
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, via Vetoio-Coppito 2, 67100 L'Aquila, Italy.
| | - Michael J Econs
- Department of Medicine, Indiana University, 1120 W. Michigan St., Indianapolis, IN 46202, USA; Department of Medical and Molecular Genetics, Indiana University, 1120 W. Michigan St., Indianapolis, IN 46202, USA.
| |
Collapse
|
9
|
Canalis E, Sanjay A, Yu J, Zanotti S. An Antibody to Notch2 Reverses the Osteopenic Phenotype of Hajdu-Cheney Mutant Male Mice. Endocrinology 2017; 158:730-742. [PMID: 28323963 PMCID: PMC5460801 DOI: 10.1210/en.2016-1787] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 01/20/2017] [Indexed: 11/19/2022]
Abstract
Notch receptors play a central role in skeletal development and bone remodeling. Hajdu-Cheney syndrome (HCS), a disease characterized by osteoporosis and fractures, is associated with gain-of-NOTCH2 function mutations. To study HCS, we created a mouse model harboring a point 6955C>T mutation in the Notch2 locus upstream of the proline, glutamic acid, serine, and threonine domain, leading to a Q2319X change at the amino acid level. Notch2Q2319X heterozygous mutants exhibited cancellous and cortical bone osteopenia. Microcomputed tomography demonstrated that the cancellous and cortical osteopenic phenotype was reversed by the administration of antibodies generated against the negative regulatory region (NRR) of Notch2, previously shown to neutralize Notch2 activity. Bone histomorphometry revealed that anti-Notch2 NRR antibodies decreased the osteoclast number and eroded surface in cancellous bone of Notch2Q2319X mice. An increase in osteoclasts on the endocortical surface of Notch2Q2319X mice was not observed in the presence of anti-Notch2 NRR antibodies. The anti-Notch2 NRR antibody decreased the induction of Notch target genes and Tnfsf11 messenger RNA levels in bone extracts and osteoblasts from Notch2Q2319X mice. In vitro experiments demonstrated increased osteoclastogenesis in Notch2Q2319X mutants in response to macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, and these effects were suppressed by the anti-Notch2 NRR. In conclusion, Notch2Q2319X mice exhibit cancellous and cortical bone osteopenia that can be corrected by the administration of anti-Notch2 NRR antibodies.
Collapse
Affiliation(s)
- Ernesto Canalis
- Departments of Orthopaedic Surgery and
- Medicine and
- the UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| | - Archana Sanjay
- Departments of Orthopaedic Surgery and
- the UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| | - Jungeun Yu
- Departments of Orthopaedic Surgery and
- the UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| | - Stefano Zanotti
- Departments of Orthopaedic Surgery and
- Medicine and
- the UConn Musculoskeletal Institute, UConn Health, Farmington, Connecticut 06030
| |
Collapse
|
10
|
Osagie-Clouard L, Sanghani A, Coathup M, Briggs T, Bostrom M, Blunn G. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017; 6:14-21. [PMID: 28062525 PMCID: PMC5227055 DOI: 10.1302/2046-3758.61.bjr-2016-0085.r1] [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: 04/10/2016] [Accepted: 10/24/2016] [Indexed: 12/19/2022] Open
Abstract
Intermittently administered parathyroid hormone (PTH 1-34) has been shown to promote bone formation in both human and animal studies. The hormone and its analogues stimulate both bone formation and resorption, and as such at low doses are now in clinical use for the treatment of severe osteoporosis. By varying the duration of exposure, parathyroid hormone can modulate genes leading to increased bone formation within a so-called 'anabolic window'. The osteogenic mechanisms involved are multiple, affecting the stimulation of osteoprogenitor cells, osteoblasts, osteocytes and the stem cell niche, and ultimately leading to increased osteoblast activation, reduced osteoblast apoptosis, upregulation of Wnt/β-catenin signalling, increased stem cell mobilisation, and mediation of the RANKL/OPG pathway. Ongoing investigation into their effect on bone formation through 'coupled' and 'uncoupled' mechanisms further underlines the impact of intermittent PTH on both cortical and cancellous bone. Given the principally catabolic actions of continuous PTH, this article reviews the skeletal actions of intermittent PTH 1-34 and the mechanisms underlying its effect. CITE THIS ARTICLE L. Osagie-Clouard, A. Sanghani, M. Coathup, T. Briggs, M. Bostrom, G. Blunn. Parathyroid hormone 1-34 and skeletal anabolic action: The use of parathyroid hormone in bone formation. Bone Joint Res 2017;6:14-21. DOI: 10.1302/2046-3758.61.BJR-2016-0085.R1.
Collapse
Affiliation(s)
- L Osagie-Clouard
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - A Sanghani
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Coathup
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - T Briggs
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| | - M Bostrom
- Hospital for Special Surgery, New York, New York, USA
| | - G Blunn
- Institute of Orthopaedics and Musculoskeletal Sciences, University College London, Royal National Orthopaedic Hospital, Stanmore, Middlesex HA7 4LP, London, UK
| |
Collapse
|
11
|
Staab E, Thiele GM, Clarey D, Wyatt TA, Romberger DJ, Wells AD, Dusad A, Wang D, Klassen LW, Mikuls TR, Duryee MJ, Poole JA. Toll-Like Receptor 4 Signaling Pathway Mediates Inhalant Organic Dust-Induced Bone Loss. PLoS One 2016; 11:e0158735. [PMID: 27479208 PMCID: PMC4968800 DOI: 10.1371/journal.pone.0158735] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 06/21/2016] [Indexed: 01/06/2023] Open
Abstract
Agriculture workers have increased rates of airway and skeletal disease. Inhalant exposure to agricultural organic dust extract (ODE) induces bone deterioration in mice; yet, mechanisms underlying lung-bone crosstalk remain unclear. Because Toll-like receptor 2 (TLR2) and TLR4 are important in mediating the airway consequences of ODE, this study investigated their role in regulating bone responses. First, swine facility ODE stimulated wild-type (WT) bone marrow macrophages to form osteoclasts, and this finding was inhibited in TLR4 knock-out (KO), but not TLR2 KO cells. Next, using an established intranasal inhalation exposure model, WT, TLR2 KO and TLR4 KO mice were treated daily with ODE or saline for 3 weeks. ODE-induced airway neutrophil influx and cytokine/chemokine release were similarly reduced in TLR2 and TLR4 KO animals as compared to WT mice. Utilizing micro-computed tomography (CT), analysis of tibia showed loss of bone mineral density, volume and deterioration of bone micro-architecture and mechanical strength induced by ODE in WT mice were significantly reduced in TLR4 but not TLR2 KO animals. Bone marrow osteoclast precursor cell populations were analyzed by flow cytometry from exposed animals. In WT animals, exposure to inhalant ODE increased osteoclast precursor cell populations as compared to saline, an effect that was reduced in TLR4 but not TLR2 KO mice. These results show that TLR2 and TLR4 pathways mediate ODE-induced airway inflammation, but bone deterioration consequences following inhalant ODE treatment is strongly dependent upon TLR4. Thus, the TLR4 signaling pathway appears critical in regulating the lung-bone inflammatory axis to microbial component-enriched organic dust exposures.
Collapse
Affiliation(s)
- Elizabeth Staab
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Geoffrey M. Thiele
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States of America
- Rheumatology Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Dillon Clarey
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Todd A. Wyatt
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States of America
- Department of Environmental, Agricultural, and Occupational Health, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Debra J. Romberger
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States of America
| | - Adam D. Wells
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Anand Dusad
- Rheumatology Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Dong Wang
- Rheumatology Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Lynell W. Klassen
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States of America
- Rheumatology Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Ted R. Mikuls
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States of America
- Rheumatology Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Michael J. Duryee
- Veterans Affairs Nebraska-Western Iowa Health Care System, Omaha, NE, United States of America
- Rheumatology Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Jill A. Poole
- Pulmonary, Critical Care, Sleep & Allergy Division, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, United States of America
- * E-mail:
| |
Collapse
|
12
|
The effects of spinal cord injury on bone loss and dysregulation of the calcium/parathyroid hormone loop in mice. Osteoporos Sarcopenia 2016; 2:164-169. [PMID: 30775482 PMCID: PMC6372742 DOI: 10.1016/j.afos.2016.06.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Revised: 06/23/2016] [Accepted: 06/27/2016] [Indexed: 11/23/2022] Open
Abstract
Objective To map the progression of osteoporosis following spinal cord injury in mice in specific areas and analyze changes in parathyroid hormone (PTH) and ion levels which could be responsible for overall bone loss. Summary of background data Spinal cord injury rapidly induces severe bone loss compared to other conditions, yet the cause of this bone loss has not been identified. Studies suggest the bone loss after injury is not solely due to disuse. Methods To quantify bone loss we weighed individual bones and measured bone mineral density using dual energy X-ray absorptiometry at acute (1 week) and chronic (4 week) time points following a T9 contusion. An ELISA was used to measure blood PTH levels at 1 and 4 weeks after injury. Calcium and phosphate levels were also analyzed at 4 weeks following injury at the University of Miami pathology core. Results We observed a significant decrease in bone mineral density in hind limbs after an acute injury, and found this bone loss to progress over time. Furthermore, following chronic injury a decrease in bone mineral density is also observed in bones above the level of injury and in the total bone mineral density. We observed a significant decrease in parathyroid hormone levels in injured mice at the chronic time point, but not at the acute time point which suggests this could be involved in the global bone loss following injury. We also observed a significant increase in serum calcium levels following injury which could account for the imbalance of PTH levels.
Collapse
|
13
|
Zhang D, Huang Y, Huang Z, Zhang R, Wang H, Huang D. FTY-720P Suppresses Osteoclast Formation by Regulating Expression of Interleukin-6 (IL-6), Interleukin-4 (IL-4), and Matrix Metalloproteinase 2 (MMP-2). Med Sci Monit 2016; 22:2187-94. [PMID: 27344392 PMCID: PMC4924886 DOI: 10.12659/msm.896690] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Background Osteoclast formation is closely related to the immune system. FTY720, a new immunosuppressive agent, has some functions in immune regulation. Its main active ingredients become FTY-720P in vivo by phosphorylation modification. The objective of this study was to determine the effects of FTY-720 with various concentrations on osteoclasts in vitro. Material/Methods RAW264.7 cells and bone marrow-derived mononuclear phagocytes (BMMs) were treated with RANKL to obtain osteoclasts in vitro. To investigate the role of FTY-720 in osteoclast formation, trap enzyme staining was performed and the number of osteoclasts was counted. Bone slices were stained with methylene blue, we counted the number of lacunae after bone slices were placed into dishes together with osteoclasts, and we observed the effect and function of FTY-720 in osteoclasts induced by RAW264.7 cells and BMMs. Then, we used a protein array kit to explore the effects of FTY-720P on osteoclasts. Results The results of enzyme trap staining and F-actin staining experiments show that, with the increasing concentration of FTY-720P, the number of osteoclast induced by RAW264.7 cells and BMMs gradually decreased (P<0.05), especially when the FTY-720P concentration reached 1000 ng/ml, and the number of osteoclasts formed was the lowest (P<0.05). With bone lacuna toluidine blue staining, the results also show that, with the increasing concentration of FTY-720P, the number of bone lacuna gradually decreased (P<0.05), and the number of lacunae is lowest when the concentration reached 800 ng/ml. Finally, protein array results showed that IL-4, IL-6, IL-12, MMP-2, VEGF-C, GFR, basic FGF, MIP-2, and insulin proteins were regulated after FTY-720P treatment. Conclusions FTY-720P can suppress osteoclast formation and function, and FTY-720P induces a series of cytokine changes.
Collapse
Affiliation(s)
- Dawei Zhang
- Section 2, Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China (mainland)
| | - Yongjun Huang
- Department of Microsurgery and Orthopedic Trauma, Guangdong No. 2 Provincial People's Hospital, Guangzhou, Guangdong, China (mainland)
| | - Zongwen Huang
- Section 2, Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China (mainland)
| | - Rongkai Zhang
- Section 2, Department of Orthopedics, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, Guangdong, China (mainland)
| | - Honggang Wang
- Department of Microsurgery and Orthopedic Trauma, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China (mainland)
| | - Dong Huang
- Department of Microsurgery and Orthopedic Trauma, Guangdong No. 2 Provincial People's Hospital, Guangzhou, Guangdong, China (mainland)
| |
Collapse
|
14
|
Oest ME, Mann KA, Zimmerman ND, Damron TA. Parathyroid Hormone (1-34) Transiently Protects Against Radiation-Induced Bone Fragility. Calcif Tissue Int 2016; 98:619-30. [PMID: 26847434 PMCID: PMC4860360 DOI: 10.1007/s00223-016-0111-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 01/18/2016] [Indexed: 01/05/2023]
Abstract
Radiation therapy for soft tissue sarcoma or tumor metastases is frequently associated with damage to the underlying bone. Using a mouse model of limited field hindlimb irradiation, we assessed the ability of parathyroid hormone (1-34) fragment (PTH) delivery to prevent radiation-associated bone damage, including loss of mechanical strength, trabecular architecture, cortical bone volume, and mineral density. Female BALB/cJ mice received four consecutive doses of 5 Gy to a single hindlimb, accompanied by daily injections of either PTH or saline (vehicle) for 8 weeks, and were followed for 26 weeks. Treatment with PTH maintained the mechanical strength of irradiated femurs in axial compression for the first eight weeks of the study, and the apparent strength of irradiated femurs in PTH-treated mice was greater than that of naïve bones during this time. PTH similarly protected against radiation-accelerated resorption of trabecular bone and transient decrease in mid-diaphyseal cortical bone volume, although this benefit was maintained only for the duration of PTH delivery. Overall, PTH conferred protection against radiation-induced fragility and morphologic changes by increasing the quantity of bone, but only during the period of administration. Following cessation of PTH delivery, bone strength and trabecular volume fraction rapidly decreased. These data suggest that PTH does not negate the longer-term potential for osteoclastic bone resorption, and therefore, finite-duration treatment with PTH alone may not be sufficient to prevent late onset radiotherapy-induced bone fragility.
Collapse
Affiliation(s)
- Megan E Oest
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA.
| | - Kenneth A Mann
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Nicholas D Zimmerman
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| | - Timothy A Damron
- Department of Orthopedic Surgery, SUNY Upstate Medical University, 750 East Adams Street, Syracuse, NY, 13210, USA
| |
Collapse
|
15
|
Choudhary S, Goetjen A, Estus T, Jacome-Galarza CE, Aguila HL, Lorenzo J, Pilbeam C. Serum Amyloid A3 Secreted by Preosteoclasts Inhibits Parathyroid Hormone-stimulated cAMP Signaling in Murine Osteoblasts. J Biol Chem 2015; 291:3882-94. [PMID: 26703472 DOI: 10.1074/jbc.m115.686576] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Indexed: 12/25/2022] Open
Abstract
Continuous parathyroid hormone (PTH) blocks its own osteogenic actions in marrow stromal cell cultures by inducing Cox2 and receptor activator of nuclear factor κB ligand (RANKL) in the osteoblastic lineage cells, which then cause the hematopoietic lineage cells to secrete an inhibitor of PTH-stimulated osteoblast differentiation. To identify this inhibitor, we used bone marrow macrophages (BMMs) and primary osteoblasts (POBs) from WT and Cox2 knock-out (KO) mice. Conditioned medium (CM) from RANKL-treated WT, but not KO, BMMs blocked PTH-stimulated cAMP production in POBs. Inhibition was reversed by pertussis toxin (PTX), which blocks Gαi/o activation. Saa3 was the most highly differentially expressed gene in a microarray comparison of RANKL-treated WT versus Cox2 KO BMMs, and RANKL induced Saa3 protein secretion only from WT BMMs. CM from RANKL-stimulated BMMs with Saa3 knockdown did not inhibit PTH-stimulated responses in POBs. SAA added to POBs inhibited PTH-stimulated cAMP responses, which was reversed by PTX. Selective agonists and antagonists of formyl peptide receptor 2 (Fpr2) suggested that Fpr2 mediated the inhibitory actions of Saa3 on osteoblasts. In BMMs committed to become osteoclasts by RANKL treatment, Saa3 expression peaked prior to appearance of multinucleated cells. Flow sorting of WT marrow revealed that Saa3 was secreted only from the RANKL-stimulated B220(-) CD3(-)CD11b(-/low) CD115(+) preosteoclast population. We conclude that Saa3 secretion from preosteoclasts, induced by RANKL in a Cox2-dependent manner, inhibits PTH-stimulated cAMP signaling and osteoblast differentiation via Gαi/o signaling. The induction of Saa3 by PTH may explain the suppression of bone formation when PTH is applied continuously and may be a new therapeutic target for osteoporosis.
Collapse
Affiliation(s)
- Shilpa Choudhary
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030 From the Departments of Medicine and
| | - Alexandra Goetjen
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030
| | - Thomas Estus
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030
| | | | | | - Joseph Lorenzo
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030 From the Departments of Medicine and
| | - Carol Pilbeam
- New England Musculoskeletal Institute, University of Connecticut Health, Farmington, Connecticut 06030 From the Departments of Medicine and
| |
Collapse
|
16
|
Canalis E, Schilling L, Yee SP, Lee SK, Zanotti S. Hajdu Cheney Mouse Mutants Exhibit Osteopenia, Increased Osteoclastogenesis, and Bone Resorption. J Biol Chem 2015; 291:1538-1551. [PMID: 26627824 DOI: 10.1074/jbc.m115.685453] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Indexed: 11/06/2022] Open
Abstract
Notch receptors are determinants of cell fate and function and play a central role in skeletal development and bone remodeling. Hajdu Cheney syndrome, a disease characterized by osteoporosis and fractures, is associated with NOTCH2 mutations resulting in a truncated stable protein and gain-of-function. We created a mouse model reproducing the Hajdu Cheney syndrome by introducing a 6955C→T mutation in the Notch2 locus leading to a Q2319X change at the amino acid level. Notch2(Q2319X) heterozygous mutants were smaller and had shorter femurs than controls; and at 1 month of age they exhibited cancellous and cortical bone osteopenia. As the mice matured, cancellous bone volume was restored partially in male but not female mice, whereas cortical osteopenia persisted in both sexes. Cancellous bone histomorphometry revealed an increased number of osteoclasts and bone resorption, without a decrease in osteoblast number or bone formation. Osteoblast differentiation and function were not affected in Notch2(Q2319X) cells. The pre-osteoclast cell pool, osteoclast differentiation, and bone resorption in response to receptor activator of nuclear factor κB ligand in vitro were increased in Notch2(Q2319X) mutants. These effects were suppressed by the γ-secretase inhibitor LY450139. In conclusion, Notch2(Q2319X) mice exhibit cancellous and cortical bone osteopenia, enhanced osteoclastogenesis, and increased bone resorption.
Collapse
Affiliation(s)
| | | | - Siu-Pok Yee
- Cell Biology, Genetics, and; Genome Sciences Biology
| | - Sun-Kyeong Lee
- Medicine,; Center on Aging, University of Connecticut Health Center, Farmington, Connecticut 06030
| | | |
Collapse
|
17
|
Jacome-Galarza C, Soung DY, Adapala NS, Pickarski M, Sanjay A, Duong LT, Lorenzo JA, Drissi H. Altered hematopoietic stem cell and osteoclast precursor frequency in cathepsin K null mice. J Cell Biochem 2015; 115:1449-57. [PMID: 24590570 DOI: 10.1002/jcb.24801] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 02/27/2014] [Indexed: 12/19/2022]
Abstract
Cathepsin K (CatK) is a lysosomal cysteine protease necessary for bone resorption by osteoclasts (OCs), which originate from myeloid hematopoietic precursors. CatK-deficient (CatK(-/-) ) mice show osteopetrosis due to defective resorption by OCs, which are increased in number in these mice. We investigated whether genetic ablation of CatK altered the number of hematopoietic stem cells (HSCs) and OC precursor cells (OCPs) using two mouse models: CatK(-/-) mice and a knock-in mouse model in which the CatK gene (ctsk) is replaced by cre recombinase. We found that CatK deletion in mice significantly increased the number of HSCs in the spleen and decreased their number in bone marrow. In contrast, the number of early OCPs was unchanged in the bone marrow. However, the number of committed CD11b(+) OCPs was increased in the bone marrow of CatK(-/-) compared to wild-type (WT) mice. In addition, the percentage but not the number of OCPs was decreased in the spleen of CatK(-/-) mice relative to WT. To understand whether increased commitment to OC lineage in CatK(-/-) mice is influenced by the bone marrow microenvironment, CatK(Cre/+) or CatK(Cre/Cre) red fluorescently labeled OCPs were injected into WT mice, which were also subjected to a mid-diaphyseal femoral fracture. The number of OCs derived from the intravenously injected CatK(Cre/Cre) OCPs was lower in the fracture callus compared to mice injected with CatK(+/Cre) OCPs. Hence, in addition to its other effects, the absence of CatK in OCP limits their ability to engraft in a repairing fracture callus compared to WT OCP.
Collapse
Affiliation(s)
- Christian Jacome-Galarza
- New England Musculoskeletal Institute, University of Connecticut Health Center, Farmington, Connecticut, 06030; Department of Medicine, University of Connecticut Health Center, Farmington, Connecticut, 06030
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Kimura S, Yoshioka K. Parathyroid hormone and parathyroid hormone type-1 receptor accelerate myocyte differentiation. Sci Rep 2014; 4:5066. [PMID: 24919035 PMCID: PMC4052750 DOI: 10.1038/srep05066] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 05/08/2014] [Indexed: 11/09/2022] Open
Abstract
The ZHTc6-MyoD embryonic stem cell line expresses the myogenic transcriptional factor MyoD under the control of a tetracycline-inducible promoter. Following induction, most of the ZHTc6-MyoD cells differentiate to myotubes. However, a small fraction does not differentiate, instead forming colonies that retain the potential for myocyte differentiation. In our current study, we found that parathyroid hormone type 1 receptor (PTH1R) expression in colony-forming cells at 13 days after differentiation was higher than that in the undifferentiated ZHTc6-MyoD cells. We also found that PTH1R expression was required for myocyte differentiation, and that parathyroid hormone accelerated the differentiation. Our analysis of human and mouse skeletal muscle tissues showed that most cells expressing PTH1R also expressed Pax7 and CD34, which are biomarkers of satellite cells. Furthermore, we found that parathyroid hormone treatment significantly improved muscle weakness in dystrophin-deficient mdx mice. This is the first report indicating that PTH1R and PTH accelerate myocyte differentiation.
Collapse
Affiliation(s)
- Shigemi Kimura
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kowasi Yoshioka
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
19
|
Šućur A, Katavić V, Kelava T, Jajić Z, Kovačić N, Grčević D. Induction of osteoclast progenitors in inflammatory conditions: key to bone destruction in arthritis. INTERNATIONAL ORTHOPAEDICS 2014; 38:1893-903. [DOI: 10.1007/s00264-014-2386-y] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 05/13/2014] [Indexed: 12/14/2022]
|
20
|
Azevedo-Neto RD, Gonzaga CC, Deliberador TM, Klug LG, Da Costa Oliveira L, Zielak JC, De Andrade Urban C, De Araujo MR, Giovanini AF. Fragmented Adipose Tissue Transplanted to Craniofacial Deformities Induces Bone Repair Associated with Immunoexpression of Adiponectin and Parathyroid Hormone 1-Receptor. Cleft Palate Craniofac J 2013; 50:639-47. [DOI: 10.1597/12-121] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Objective This study analyzed the influence of autogenous white adipose tissue on bone matrix development in critical-size defects created in rabbit calvaria. Materials and Methods A 15-mm-diameter defect was created in the calvaria of 42 rabbits. Twenty-one rabbits were treated with 86 mm3 of immediate transplant of fragmented white subcutaneous adipose tissue (WSAT); the others constituted the control group (sham). The animals were euthanized at 7, 15, and 40 days postsurgery (n = 7), and the histological data were analyzed by histomorphometry and immunohistochemistry using the anti-adiponectin and parathyroid hormone 1-receptor (PTH1R) antibodies. Results The calvariae treated with fragmented WSAT demonstrated significant bone formation. These results coincided with the significant presence of immunopositivity to adiponectin and PTH1R in loci, which in turn coincided with the increase in bonelike matrix deposited both in fat tissue stroma and adipocytes' cytoplasm. In contrast, the control group revealed a small amount of bone-matrix deposition and presented scarce PTH1R expression and a lack of immunostain for adiponectin. Conclusion These results indicate that transplant of fragmented white subcutaneous adipose tissue may be an alternative to treatment of craniofacial bone deformities because adipose tissue suffers from osseous metaplasia and exhibits immunoexpression of the adiponectin and PTH1R, which are proteins associated with bone metabolism
Collapse
|
21
|
Therapeutic effect of aqueous extract from Ecliptae herba on bone metabolism of ovariectomized rats. Menopause 2013; 20:232-40. [PMID: 23096243 DOI: 10.1097/gme.0b013e318265e7dd] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
OBJECTIVE Ecliptae herba (EH) has long been used in China to strengthen bones. Accumulating evidence indicates that EH may have antiosteoporotic effects. The aim of this study was to evaluate the effects of aqueous EH extract (EHE) on rats that had osteoporosis-like features induced by ovariectomy, using aqueous Fructus Ligustri Lucidi extract as positive control agent. METHODS Three-month-old female rats that underwent ovariectomy were treated with EHE (1.4 g/kg per day). After 12 weeks, bone mineral density and bone histomorphometric indices of tibiae were measured. Protein and messenger RNA expressions of osteoprotegerin and receptor activator of nuclear factor κ-B ligand (RANKL) in tibiae were evaluated by immunohistochemistry and in situ hybridization. In addition, serum concentrations of osteocalcin, interleukin-1β, interleukin-6 (IL-6), calcitonin (CT), and parathyroid hormone were determined by enzyme-linked immunosorbent assay. RESULTS EHE treatment prevented body weight gain and loss of uterine wet weight in ovariectomized rats. It remarkably increased bone mass in ovariectomized rats compared with ovariectomized controls. EHE treatment significantly down-regulated RANKL expression in tibiae from ovariectomized rats compared with controls; however, it had no significant effect on osteoprotegerin expression. In addition, EHE treatment significantly reduced serum IL-6 levels and remarkably increased CT levels but had no effect on parathyroid hormone. CONCLUSIONS EHE increases bone mass in ovariectomized rats by inhibiting bone loss: down-regulated RANKL expression in tibiae and IL-6 level in serum, and up-regulated CT level in serum. This suggests that EHE may be developed as an alternative therapeutic agent for osteoporosis induced by postmenopause.
Collapse
|
22
|
Kuroshima S, Elliott KW, Yamashita J. Effect of zoledronate on the responses of osteocytes to acute parathyroid hormone. Calcif Tissue Int 2013; 92:576-85. [PMID: 23503790 PMCID: PMC3656166 DOI: 10.1007/s00223-013-9720-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Accepted: 01/29/2013] [Indexed: 01/09/2023]
Abstract
The bone anabolic effect of parathyroid hormone (PTH) therapy is blunted when used in patients who were previously on bisphosphonate treatment. Osteocytes may play a role in the bisphosphonate silencing effect on PTH therapy since bisphosphonates have been shown to reach the lacunocanalicular system. In vivo osteocyte studies pose a significant challenge. For the current study, we developed a simple method to isolate RNA from cortical bone enriched with osteocytes. Our purpose was to investigate how zoledronate (ZA) treatment modulates the responses of osteocytes and the bone marrow (BM) to acute PTH treatment. Mice received ZA treatment for 3 months and a single PTH injection prior to death. Bone was histomorphometrically evaluated. Gene expression was assessed at the RNA level in osteocytes and BM. Endothelial progenitor cells (EPCs) and γδT cells were analyzed in the BM and blood using flow cytometry. We found that ZA treatment altered bone responses to PTH. Expression of Sfrp4, a Wnt antagonist, was significantly increased in ZA-affected osteocytes. BM EPCs were increased in response to acute PTH but not when treatment was combined with ZA. ZA treatment augmented EPCs in the BM but not in blood, which suggests that ZA treatment may have differential effects between the BM and blood. These findings indicate that osteocytes and BM EPCs in mice on ZA treatment respond differently to acute PTH from those not receiving ZA. This may partially explain the mechanisms of previous reports that ZA therapy attenuates the anabolic effect of PTH in bone.
Collapse
Affiliation(s)
- Shinichiro Kuroshima
- Department of Biologic and Materials Sciences, School of Dentistry, University of Michigan, 1011 North University Avenue, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
23
|
Therapeutic Effects of Cortex acanthopanacis Aqueous Extract on Bone Metabolism of Ovariectomized Rats. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:492627. [PMID: 22997530 PMCID: PMC3446781 DOI: 10.1155/2012/492627] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2012] [Accepted: 08/10/2012] [Indexed: 12/01/2022]
Abstract
The aim of this study was to evaluate effects of aqueous extract from Cortex acanthopanacis (CAE) on osteoporosis rats induced by ovariectomy (OVX) using aqueous extract from Folium Epimedii (FEE) as positive control agent. Three-month-old female rats that underwent OVX were treated with CAE. After 12 weeks, bone mineral density (BMD) and indices of bone histomorphometry of tibia were measured. Levels of protein and mRNA expression of osteoprotegerin (OPG) and receptor activator of nuclear factor kappa-B ligand (RANKL) in tibia were evaluated. In addition, the serum concentrations of osteocalcin (OC), interleukin-1 beta (IL-1β), interleukin-6 (IL-6), calcitonin (CT), and parathyroid hormone (PTH) were determined. Administration of CAE significantly prevented OVX-induced rats from gain of the body weight. Treatment with CAE increased bone mass remarkably and showed a significant inhibitory effect on bone resorption by downregulating significantly the expression of RANKL in tibia of OVX rats. Meanwhile, treatment of CAE significantly reduced serum level of IL-1β and increased level of CT in OVX rats. This suggests that CAE has the potential to be used as an alternative therapeutic agent for postmenopausal osteoporosis.
Collapse
|
24
|
Ketteler M, Martin KJ, Wolf M, Amdahl M, Cozzolino M, Goldsmith D, Sharma A, Marx S, Khan S. Paricalcitol versus cinacalcet plus low-dose vitamin D therapy for the treatment of secondary hyperparathyroidism in patients receiving haemodialysis: results of the IMPACT SHPT study. Nephrol Dial Transplant 2012; 27:3270-8. [PMID: 22387567 PMCID: PMC3408938 DOI: 10.1093/ndt/gfs018] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Optimal treatment for secondary hyperparathyroidism (SHPT) has not been defined. The IMPACT SHPT (ClinicalTrials.gov identifier: NCT00977080) study assessed whether dose-titrated paricalcitol plus supplemental cinacalcet only for hypercalcaemia is superior to cinacalcet plus low-dose vitamin D in controlling intact parathyroid hormone (iPTH) levels in patients with SHPT on haemodialysis. METHODS In this 28-week, multicentre, open-label Phase 4 study, participants were randomly selected to receive paricalcitol or cinacalcet plus low-dose vitamin D. Randomization and analyses were stratified by mode of paricalcitol administration [intravenous (IV) or oral]. The primary efficacy end point was the proportion of subjects who achieved a mean iPTH value of 150-300 pg/mL during Weeks 21-28. RESULTS Of 272 subjects randomized, 268 received one or more dose of study drug; 101 in the IV and 110 in the oral stratum with two or more values during Weeks 21-28 were included in the primary analysis. In the IV stratum, 57.7% of subjects in the paricalcitol versus 32.7% in the cinacalcet group (P = 0.016) achieved the primary end point. In the oral stratum, the corresponding proportions of subjects were 54.4% for paricalcitol and 43.4% for cinacalcet (P = 0.260). Cochran-Mantel-Haenszel analysis, controlling for stratum, revealed overall superiority of paricalcitol (56.0%) over cinacalcet (38.2%; P = 0.010) in achieving iPTH 150-300 pg/mL during Weeks 21-28. Hypercalcaemia occurred in 4 (7.7%) and 0 (0%) of paricalcitol-treated subjects in the IV and oral strata, respectively. Hypocalcaemia occurred in 46.9% and 54.7% of cinacalcet-treated subjects in the IV and oral strata, respectively. CONCLUSION Paricalcitol versus cinacalcet plus low-dose vitamin D provided superior control of iPTH, with low incidence of hypercalcaemia.
Collapse
|
25
|
Mercier FE, Ragu C, Scadden DT. The bone marrow at the crossroads of blood and immunity. Nat Rev Immunol 2011; 12:49-60. [PMID: 22193770 DOI: 10.1038/nri3132] [Citation(s) in RCA: 238] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Progenitor cells that are the basis for all blood cell production share the bone marrow with more mature elements of the adaptive immune system. Specialized niches within the bone marrow guide and, at times, constrain the development of haematopoietic stem and progenitor cells (HSPCs) and lineage-restricted immune progenitor cells. Specific niche components are organized into distinct domains to create a diversified landscape in which specialized cell differentiation or population expansion programmes proceed. Local cues that reflect the tissue and organismal state affect cellular interactions to alter the production of a range of cell types. Here, we review the organization of regulatory elements in the bone marrow and discuss how these elements provide a dynamic means for the host to modulate stem cell and adaptive immune cell responses to physiological challenges.
Collapse
Affiliation(s)
- Francois E Mercier
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Christine Ragu
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - David T Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
26
|
Mercier FE, Ragu C, Scadden DT. The bone marrow at the crossroads of blood and immunity. Nat Rev Immunol 2011. [PMID: 22193770 DOI: 10.1038/nri4132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Progenitor cells that are the basis for all blood cell production share the bone marrow with more mature elements of the adaptive immune system. Specialized niches within the bone marrow guide and, at times, constrain the development of haematopoietic stem and progenitor cells (HSPCs) and lineage-restricted immune progenitor cells. Specific niche components are organized into distinct domains to create a diversified landscape in which specialized cell differentiation or population expansion programmes proceed. Local cues that reflect the tissue and organismal state affect cellular interactions to alter the production of a range of cell types. Here, we review the organization of regulatory elements in the bone marrow and discuss how these elements provide a dynamic means for the host to modulate stem cell and adaptive immune cell responses to physiological challenges.
Collapse
Affiliation(s)
- Francois E Mercier
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - Christine Ragu
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| | - David T Scadden
- Center for Regenerative Medicine and Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA.,Harvard Stem Cell Institute and Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, Massachusetts 02138, USA
| |
Collapse
|
27
|
Proteoglycan 4, a novel immunomodulatory factor, regulates parathyroid hormone actions on hematopoietic cells. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2431-42. [PMID: 21939632 PMCID: PMC3204095 DOI: 10.1016/j.ajpath.2011.07.032] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2011] [Revised: 06/12/2011] [Accepted: 07/07/2011] [Indexed: 01/10/2023]
Abstract
Proteoglycan 4 (PRG4), a critical protective factor in articular joints, is implicated in hematopoietic progenitor cell expansion and megakaryopoiesis. PRG4 loss-of-function mutations result in camptodactyly-arthropathy-coxa vara-pericarditis (CACP) syndrome, which is characterized primarily by precocious joint failure. PRG4 was identified as a novel parathyroid hormone (PTH) responsiveness gene in osteoblastic cells in bone, and was investigated as a potential mediator of PTH actions on hematopoiesis. Sixteen-week-old Prg4(-/-) mutant and Prg4(+/+) wild-type mice were treated daily with intermittent PTH (residues 1-34) or vehicle for 6 weeks. At 22 weeks of age, Prg4 mutant mice had increased peripheral blood neutrophils and decreased marrow B220(+) (B-lymphocytic) cells, which were normalized by PTH. The PTH-induced increase in marrow Lin(-)Sca-1(+)c-Kit(+) (hematopoietic progenitor) cells was blunted in mutant mice. Basal and PTH-stimulated stromal cell-derived factor-1 (SDF-1) was decreased in mutant mice, suggesting SDF-1 as a candidate regulator of proteoglycan 4 actions on hematopoiesis in vivo. PTH stimulation of IL-6 mRNA was greater in mutant than in wild-type calvaria and bone marrow, suggesting a compensatory mechanism in the PTH-induced increase in marrow hematopoietic progenitor cells. In summary, proteoglycan 4 is a novel PTH-responsive factor regulating immune cells and PTH actions on marrow hematopoietic progenitor cells.
Collapse
|