1
|
Shao C, Chen H, Liu T, Pan C. The Hippo pathway in bone and cartilage: implications for development and disease. PeerJ 2025; 13:e19334. [PMID: 40292098 PMCID: PMC12024444 DOI: 10.7717/peerj.19334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025] Open
Abstract
Bone is the main structure of the human body; it mainly plays a supporting role and participates in metabolic processes. The Hippo signaling pathway is composed of a series of protein kinases, including the mammalian STE20-like kinase MST1/2 and the large tumor suppressor LATS1/2, which are widely involved in pathophysiological processes, including cell proliferation, differentiation, apoptosis and death, especially those related to biomechanical transduction in vivo. However, the role of it in regulating skeletal system development and the evolution of bone-related diseases remains poorly understood. The pathway can intervene in and regulate the physiological activities of bone-related cells such as osteoclasts and chondrocytes through its own or other bone-related signaling pathways, such as the Wnt pathway, the Notch pathway, and receptor activator of nuclear factor-κB ligand (RANKL), thereby affecting the occurrence and development of bone diseases. This article discusses the role of the Hippo signaling pathway in bone development and disease to provide new insights into the treatment of bone-related diseases by targeting the Hippo signaling pathway.
Collapse
Affiliation(s)
- Chenwei Shao
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Hao Chen
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Tingting Liu
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| | - Chun Pan
- Institute of Translational Medicine, Yangzhou University, Yangzhou, Jiangsu, China
- Department of Orthopedics, Affiliated Hospital of Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
2
|
Xiang Q, Li L, Ji W, Gawlitta D, Walboomers XF, van den Beucken JJJP. Beyond resorption: osteoclasts as drivers of bone formation. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:22. [PMID: 39392536 PMCID: PMC11469995 DOI: 10.1186/s13619-024-00205-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Emerging evidence illustrates that osteoclasts (OCs) play diverse roles beyond bone resorption, contributing significantly to bone formation and regeneration. Despite this, OCs remain mysterious cells, with aspects of their lifespan-from origin, fusion, alterations in cellular characteristics, to functions-remaining incompletely understood. Recent studies have identified that embryonic osteoclastogenesis is primarily driven by osteoclast precursors (OCPs) derived from erythromyeloid progenitors (EMPs). These precursor cells subsequently fuse into OCs essential for normal bone development and repair. Postnatally, hematopoietic stem cells (HSCs) become the primary source of OCs, gradually replacing EMP-derived OCs and assuming functional roles in adulthood. The absence of OCs during bone development results in bone structure malformation, including abnormal bone marrow cavity formation and shorter long bones. Additionally, OCs are reported to have intimate interactions with blood vessels, influencing bone formation and repair through angiogenesis regulation. Upon biomaterial implantation, activation of the innate immune system ensues immediately. OCs, originating from macrophages, closely interact with the immune system. Furthermore, evidence from material-induced bone formation events suggests that OCs are pivotal in these de novo bone formation processes. Nevertheless, achieving a pure OC culture remains challenging, and interpreting OC functions in vivo faces difficulties due to the presence of other multinucleated cells around bone-forming biomaterials. We here describe the fusion characteristics of OCPs and summarize reliable markers and morphological changes in OCs during their fusion process, providing guidance for researchers in identifying OCs both in vitro and in vivo. This review focuses on OC formation, characterization, and the roles of OCs beyond resorption in various bone pathophysiological processes. Finally, therapeutic strategies targeting OCs are discussed.
Collapse
Affiliation(s)
- Qianfeng Xiang
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
| | - Lei Li
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Wei Ji
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Debby Gawlitta
- Department of Oral and Maxillofacial Surgery & Special Dental Care, University Medical Center Utrecht, Utrecht University, Utrecht, GA, 3508, The Netherlands
- Regenerative Medicine Center Utrecht, Utrecht, CT, 3584, The Netherlands
| | - X Frank Walboomers
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands
- Research Institute for Medical Innovation, Radboudumc, Nijmegen, the Netherlands
| | - Jeroen J J P van den Beucken
- Radboudumc, Dentistry - Regenerative Biomaterials, Philips Van Leijdenlaan 25, Nijmegen, 6525EX, the Netherlands.
| |
Collapse
|
3
|
Harrison K, Loundagin L, Hiebert B, Panahifar A, Zhu N, Marchiori D, Arnason T, Swekla K, Pivonka P, Cooper D. Glucocorticoids disrupt longitudinal advance of cortical bone basic multicellular units in the rabbit distal tibia. Bone 2024; 187:117171. [PMID: 38901788 DOI: 10.1016/j.bone.2024.117171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 06/13/2024] [Accepted: 06/15/2024] [Indexed: 06/22/2024]
Abstract
Glucocorticoids (GCs) are the leading cause of secondary osteoporosis. The emerging perspective, derived primarily from 2D histological study of trabecular bone, is that GC-induced bone loss arises through the uncoupling of bone formation and resorption at the level of the basic multicellular unit (BMU), which carries out bone remodeling. Here we explore the impact of GCs on cortical bone remodeling in the rabbit model. Based upon the rapid reduction of bone formation and initial elevation of resorption caused by GCs, we hypothesized that the rate of advance (longitudinal erosion rate; LER) of cortical BMUs would be increased. To test this hypothesis we divided 20 female New Zealand White rabbits into four experimental groups: ovariohysterectomy (OVH), glucocorticoid (GC), OVH + GC and SHAM controls (n = 5 animals each). Ten weeks post-surgery (OVH or sham), and two weeks after the initiation of dosing (daily subcutaneous injections of 1.5 mg/kg of methylprednisolone sodium succinate in the GC-treated groups and 1 ml of saline for the others), the right tibiae were scanned in vivo using Synchrotron Radiation (SR) in-line phase contrast micro-CT at the Canadian Light Source. After an additional 2 weeks of dosing, the rabbits were euthanized and ex vivo images were collected using desktop micro-CT. The datasets were co-registered in 3D and LER was calculated as the distance traversed by BMU cutting-cones in the 14-day interval between scans. Counter to our hypothesis, LER was greatly reduced in GC-treated rabbits. Mean LER was lower in GC (4.27 μm/d; p < 0.001) and OVH + GC (4.19 μm/d; p < 0.001), while similar in OVH (40.13 μm/d; p = 0.990), compared to SHAM (40.44 μm/d). This approximately 90 % reduction in LER with GCs was also associated with an overall disruption of BMU progression, with radial expansion of the remodeling space occurring in all directions. This unexpected outcome suggests that GCs do not simply uncouple formation and resorption within cortical BMUs and highlights the value of the time-lapsed 4D approach employed.
Collapse
Affiliation(s)
- Kim Harrison
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Lindsay Loundagin
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Beverly Hiebert
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada; Max Rady College of Medicine, University of Manitoba, Winnipeg, Canada
| | - Arash Panahifar
- BioMedical Imaging and Therapy Beamline, Canadian Light Source, Saskatoon, Canada; Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Ning Zhu
- BioMedical Imaging and Therapy Beamline, Canadian Light Source, Saskatoon, Canada; Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, Saskatoon, Canada
| | - Denver Marchiori
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Terra Arnason
- Medicine Dept of Endocrinology, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, Canada
| | - Kurtis Swekla
- Animal Care and Research Support Office, Office of the Vice President of Research, University of Saskatchewan, Saskatoon, Canada
| | - Peter Pivonka
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, Australia
| | - David Cooper
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada.
| |
Collapse
|
4
|
Loundagin LL, Harrison KD, Wei X, Cooper DML. Understanding basic multicellular unit activity in cortical bone through 3D morphological analysis: New methods to define zones of the remodeling space. Bone 2024; 179:116960. [PMID: 37972746 DOI: 10.1016/j.bone.2023.116960] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/27/2023] [Accepted: 11/08/2023] [Indexed: 11/19/2023]
Abstract
The activity of basic multicellular units (BMU) in cortical bone is classically described as a sequential order of events- resorption, reversal and formation. This simplified portrayal of the remodeling process is pervasive despite the reported variability in remodeling space morphology. These variations may reflect meaningful nuances in BMU activity but methods to quantify 3D remodeling space morphology within the context of the cellular activity are currently lacking. This study developed new techniques to define zones of BMU activity based on the 3D morphology of remodeling spaces in rabbit cortical bone and integrated morphological data with the BMU longitudinal erosion rate (LER) to elucidate the spatial-temporal coordination of BMUs and estimate mineral apposition rate (MAR). The tibiae of New Zealand white rabbits (n = 5) were imaged in vivo using synchrotron radiation and two weeks later ex vivo with desktop microCT. The in vivo and ex vivo datasets were co-registered, and 27 remodeling spaces were identified at both timepoints. A radial profile representing the 3D morphology was the platform for partitioning the remodeling spaces into resorption, reversal and formation zones. Manual, automated and semi-automated partitioning approaches were compared, and the zone-segmentations were used to calculate the length, change in radius and slope of each zone. The manual approach most accurately defined the zones of idealized remodeling spaces with known dimensions (relative error = 0.9-9.2 %) while the semi-automated method reliably defined the zones in rabbit remodeling spaces (ICC = 0.85-1.00). Combining LER and the manually derived zone dimensions indicated that a BMU passes through a cross-section in approximately 18.8 days with resorption, reversal and formation taking 4.1, 2.2, and 12.5 days, respectively. MAR estimated by the 3D analysis was not significantly different than that determined with classic histomorphometry (p = 0.48). These techniques have the potential to assess dynamic parameters of bone resorption and formation, eliminate the need for fluorochrome labeling and provide a more comprehensive perspective of the remodeling process.
Collapse
Affiliation(s)
- Lindsay L Loundagin
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada.
| | - Kim D Harrison
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Xuan Wei
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - David M L Cooper
- Department of Anatomy, Physiology and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
5
|
van Dijk Christiansen P, Andreasen CM, El-Masri BM, Laursen KS, Delaisse JM, Andersen TL. Osteoprogenitor recruitment and differentiation during intracortical bone remodeling of adolescent humans. Bone 2023; 177:116896. [PMID: 37699496 DOI: 10.1016/j.bone.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 08/17/2023] [Accepted: 09/08/2023] [Indexed: 09/14/2023]
Abstract
BACKGROUND Recruitment and proliferation of osteoprogenitors during the reversal-resorption phase, and their differentiation into mature bone-forming osteoblasts is crucial for initiation of bone formation during bone remodeling. This study investigates the osteoprogenitors' gradual recruitment, proliferation, and differentiation into bone-forming osteoblasts within intracortical remodeling events of healthy adolescent humans. METHODS The study was conducted on cortical bone specimens from 11 adolescent human controls - patients undergoing surgery due to coxa valga. The osteoprogenitor recruitment route and differentiation into osteoblasts were backtracked using immunostainings and in situ hybridizations with osteoblastic markers (CD271/NGFR, osterix/SP7, COL3A1 and COL1A1). The osteoblastic cell populations were defined based on the pore surfaces, and their proliferation index (Ki67), density and number/circumference were estimated in multiplex-immunofluorescence (Ki67, TRAcP, CD34) stained sections. RESULTS During the reversal-resorption phase, osteoclasts are intermixed with (COL3A1+NFGR+) osteoblastic reversal cells, which are considered to be osteoprogenitors of (COL1A1+SP7+) bone-forming osteoblasts. Initiation of bone formation requires a critical density of these osteoprogenitors (43 ± 9 cells/mm), which is reached though proliferation (4.4 ± 0.5 % proliferative) and even more so through recruitment of osteoprogenitors, but challenged by the ongoing expansion of the canal circumference. These osteoprogenitors most likely originate from osteoblastic bone lining cells and mainly lumen osteoprogenitors, which expand their population though proliferation (4.6 ± 0.3 %) and vascular recruitment. These lumen osteoprogenitors resemble canopy cells above trabecular remodeling sites, and like canopy cells they extend above bone-forming osteoblasts where they may rejuvenate the osteoblast population during bone formation. CONCLUSION Initiation of bone formation during intracortical remodeling requires a critical density of osteoprogenitors on eroded surfaces, which is reached though proliferation and recruitment of local osteoprogenitors: bone lining cells and lumen osteoprogenitors.
Collapse
Affiliation(s)
- Pernille van Dijk Christiansen
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
| | - Christina Møller Andreasen
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| | - Bilal Mohamad El-Masri
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| | - Kaja Søndergaard Laursen
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| | - Jean-Marie Delaisse
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Thomas Levin Andersen
- Clinical Cell Biology, Research Unit of Pathology, Department of Pathology, Odense University Hospital and Department of Clinical Research, University of Southern Denmark, Odense, Denmark; Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Forensic Medicine, Aarhus University, Aarhus, Denmark; Danish Spatial Imaging Consortium (DanSIC).
| |
Collapse
|
6
|
Drejer LA, El-Masri BM, Ejersted C, Andreasen CM, Thomsen LK, Thomsen JS, Andersen TL, Hansen S. Trabecular bone deterioration in a postmenopausal female suffering multiple spontaneous vertebral fractures due to a delayed denosumab injection - A post-treatment re-initiation bone biopsy-based case study. Bone Rep 2023; 19:101703. [PMID: 37576928 PMCID: PMC10412862 DOI: 10.1016/j.bonr.2023.101703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/12/2023] [Accepted: 07/20/2023] [Indexed: 08/15/2023] Open
Abstract
Background Denosumab, is a potent anti-resorptive that, increases bone mineral density, and reduces fracture risk in osteoporotic patients. However, several case studies have reported multiple vertebral fractures in patients discontinuing denosumab. Case presentation This case report describes a 64-year-old female with postmenopausal osteoporosis treated with denosumab, who had her 11th injection delayed by 4 months. The patient suffered eight spontaneous vertebral fractures. After consent, an iliac crest bone biopsy was obtained following re-initiation of the denosumab treatment and analyzed by micro-computed tomography and histomorphometry. Results micro-computed tomography analysis revealed a low trabecular bone volume of 10 %, a low trabecular thickness of 97 μm, a low trabecular spacing of 546 μm, a high trabecular number of 1.8/mm, and a high structure model index of 2.2, suggesting trabecular thinning and loss of trabecular plates. Histomorphometric trabecular bone analysis revealed an eroded perimeter per bone perimeter of 33 % and an osteoid perimeter per bone perimeter of 62 %. Importantly, 88 % of the osteoid perimeter was immediately above an eroded-scalloped cement line with no sign of mineralization, and often with no clear bone-forming osteoblasts on the surface. Moreover, only 5 % of the bone perimeter was mineralizing, reflecting that only 8 % of the osteoid perimeter underwent mineralization, resulting in a mineralization lag time of 545 days. Taken together, this indicates limited bone formation and delayed mineralization. Conclusion We present a case report of multiple vertebral fractures after denosumab discontinuation with histomorphometric evidence that denosumab discontinuation leads to extensive trabecular bone resorption followed by a limited bone formation and delayed mineralization if the denosumab treatment is reinitiated. This highlights the importance of developing optimal discontinuation strategies for patients that are to discontinue treatment.
Collapse
Affiliation(s)
- Louise Alstrup Drejer
- Department of Endocrinology, University Hospital of Southern Denmark, Esbjerg, Denmark
| | - Bilal Mohamad El-Masri
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Charlotte Ejersted
- Department of Endocrinology, Odense University Hospital, Odense, Denmark
| | - Christina Møller Andreasen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Lisbeth Koch Thomsen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | - Thomas Levin Andersen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| | - Stinus Hansen
- Department of Endocrinology, University Hospital of Southern Denmark, Esbjerg, Denmark
| |
Collapse
|
7
|
van Dijk Christiansen P, Sikjær T, Andreasen CM, Thomsen JS, Brüel A, Hauge EM, Delaisse J, Rejnmark L, Andersen TL. Transitory Activation and Improved Transition from Erosion to Formation within Intracortical Bone Remodeling in Hypoparathyroid Patients Treated with rhPTH(1-84). JBMR Plus 2023; 7:e10829. [PMID: 38130746 PMCID: PMC10731115 DOI: 10.1002/jbm4.10829] [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: 08/13/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 12/23/2023] Open
Abstract
In hypoparathyroidism, lack of parathyroid hormone (PTH) leads to low calcium levels and decreased bone remodeling. Treatment with recombinant human PTH (rhPTH) may normalize bone turnover. This study aimed to investigate whether rhPTH(1-84) continued to activate intracortical bone remodeling after 30 months and promoted the transition from erosion to formation and whether this effect was transitory when rhPTH(1-84) was discontinued. Cortical histomorphometry was performed on 60 bone biopsies from patients (aged 31 to 78 years) with chronic hypoparathyroidism randomized to either 100 μg rhPTH(1-84) a day (n = 21) (PTH) or similar placebo (n = 21) (PLB) for 6 months as add-on to conventional therapy. This was followed by an open-label extension, where patients extended their rhPTH(1-84) (PTH) (n = 5), continued conventional treatment (CON) (n = 5), or withdrew from rhPTH(1-84) and resumed conventional therapy (PTHw) for an additional 24 months (n = 8). Bone biopsies were collected at months 6 (n = 42) and 30 (n = 18). After 6 and 30 months, the overall cortical microarchitecture (cortical porosity, thickness, pore density, and mean pore diameter) in the PTH group did not differ from that of the PLB/CON and PTHw groups. Still, the PTH group had a significantly and persistently higher percentage of pores undergoing remodeling than the PLB/CON groups. A significantly higher percentage of these pores was undergoing bone formation in the PTH compared with the PLB/CON groups, whereas the percentage of pores with erosion only was not different. This resulted in a shift in the ratio between formative and eroded pores, reflecting a faster transition from erosion to formation in the PTH-treated patients. In the rhPTH(1-84) withdrawal group PTHw, the latter effects of PTH were completely reversed in comparison to those of the PLB/CON groups. In conclusion, rhPTH(1-84) replacement therapy in hypoparathyroidism patients promotes intracortical remodeling and its transition from erosion to formation without affecting the overall cortical microstructure. The effect persists for at least 30 months and is reversible when treatment is withdrawn. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Pernille van Dijk Christiansen
- Department of PathologyOdense University HospitalOdenseDenmark
- Molecular Bone Histology (MBH) Lab, Research Unit of Pathology, Department of Clinical Research and Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Tanja Sikjær
- Department of Endocrinology and Internal MedicineAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Christina Møller Andreasen
- Department of PathologyOdense University HospitalOdenseDenmark
- Molecular Bone Histology (MBH) Lab, Research Unit of Pathology, Department of Clinical Research and Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | | | | | - Ellen Margrethe Hauge
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
- Department of RheumatologyAarhus University HospitalAarhusDenmark
| | - Jean‐Marie Delaisse
- Department of PathologyOdense University HospitalOdenseDenmark
- Molecular Bone Histology (MBH) Lab, Research Unit of Pathology, Department of Clinical Research and Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Lars Rejnmark
- Department of Endocrinology and Internal MedicineAarhus University HospitalAarhusDenmark
- Department of Clinical MedicineAarhus UniversityAarhusDenmark
| | - Thomas Levin Andersen
- Department of PathologyOdense University HospitalOdenseDenmark
- Molecular Bone Histology (MBH) Lab, Research Unit of Pathology, Department of Clinical Research and Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- Molecular Bone Histology (MBH) Lab, Department of Forensic MedicineAarhus UniversityAarhusDenmark
| |
Collapse
|
8
|
Sørensen NN, Andreasen CM, Jensen PR, Hauge EM, Bollerslev J, Delaissé JM, Kassem M, Jafari A, Diaz-delCastillo M, Andersen TL. Disturbed bone marrow adiposity in patients with Cushing's syndrome and glucocorticoid- and postmenopausal- induced osteoporosis. Front Endocrinol (Lausanne) 2023; 14:1232574. [PMID: 37881495 PMCID: PMC10597666 DOI: 10.3389/fendo.2023.1232574] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/19/2023] [Indexed: 10/27/2023] Open
Abstract
Background Skeletal stem/progenitor cells (SSPCs) in the bone marrow can differentiate into osteoblasts or adipocytes in response to microenvironmental signalling input, including hormonal signalling. Glucocorticoids (GC) are corticosteroid hormones that promote adipogenic differentiation and are endogenously increased in patients with Cushing´s syndrome (CS). Here, we investigate bone marrow adiposity changes in response to endogenous or exogenous GC increases. For that, we characterize bone biopsies from patients with CS and post-menopausal women with glucocorticoid-induced osteoporosis (GC-O), compared to age-matched controls, including postmenopausal osteoporotic patients (PM-O). Methods Transiliac crest bone biopsies from CS patients and healthy controls, and from postmenopausal women with GC-O and matched controls were analysed; an additional cohort included biopsies from women with PM-O. Plastic-embedded biopsies were sectioned for histomorphometric characterization and quantification of adipocytes. The fraction of adipocyte area per tissue (Ad.Ar/T.Ar) and marrow area (Ad.Ar/Ma.Ar), mean adipocyte profile area (Ad.Pf.Ar) and adipocyte profile density (N.Ad.Pf/Ma.Ar) were determined and correlated to steroid levels. Furthermore, the spatial distribution of adipocytes in relation to trabecular bone was characterized and correlations between bone marrow adiposity and bone remodeling parameters investigated. Results Biopsies from patients with CS and GC-O presented increased Ad.Ar/Ma.Ar, along with adipocyte hypertrophy and hyperplasia. In patients with CS, both Ad.Ar/Ma.Ar and Ad.Pf.Ar significantly correlated with serum cortisol levels. Spatial distribution analyses revealed that, in CS, the increase in Ad.Ar/Ma.Ar near to trabecular bone (<100 µm) was mediated by both adipocyte hypertrophy and hyperplasia, while N.Ad.Pf/Ma.Ar further into the marrow (>100 µm) remained unchanged. In contrast, patients with GC-O only presented increased Ad.Ar/Ma.Ar and mean Ad.Pf.Ar>100 µm from trabecular bone surface, highlighting the differential effect of increased endogenous steroid accumulation. Finally, the Ad.Ar/Ma.Ar and Ad.Ar/T.Ar correlated with the canopy coverage above remodeling events. Conclusion Increased cortisol production in patients with CS induces increased bone marrow adiposity, primarily mediated by adipocyte hypertrophy. This adiposity is particularly evident near trabecular bone surfaces, where hyperplasia also occurs. The differential pattern of adiposity in patients with CS and GC-O highlights that bone marrow adipocytes and their progenitors may respond differently in these two GC-mediated bone diseases.
Collapse
Affiliation(s)
- Nina N. Sørensen
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Christina M. Andreasen
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Danish Spatial Imaging Consortium (DanSIC), Denmark
| | - Pia R. Jensen
- Clinical Cell Biology (KCB), Vejle/Lillebaelt Hospital, Institute of Regional Health Research (IRS), University of Southern Denmark, Vejle, Denmark
| | - Ellen M. Hauge
- Department of Rheumatology, Aarhus University Hospital, Aarhus, Denmark
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Oslo University Hospital, Oslo, Norway
- Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jean-Marie Delaissé
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Moustapha Kassem
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Abbas Jafari
- Department of Cellular and Molecular Medicine, Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark
- Molecular Endocrinology & Stem Cell Research Unit (KMEB), Department of Endocrinology and Metabolism, Odense University Hospital & University of Southern Denmark, Odense, Denmark
| | - Marta Diaz-delCastillo
- Danish Spatial Imaging Consortium (DanSIC), Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| | - Thomas L. Andersen
- Research Unit of Pathology, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Danish Spatial Imaging Consortium (DanSIC), Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
9
|
Everts V, Jansen IDC, de Vries TJ. Mechanisms of bone resorption. Bone 2022; 163:116499. [PMID: 35872106 DOI: 10.1016/j.bone.2022.116499] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/12/2022] [Accepted: 07/15/2022] [Indexed: 01/08/2023]
Affiliation(s)
- Vincent Everts
- Department of Oral Cell Biology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, Amsterdam, the Netherlands; Department of Anatomy, Dental Faculty, Chulalongkorn University, Bangkok, Thailand.
| | - Ineke D C Jansen
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, Amsterdam, the Netherlands
| | - Teun J de Vries
- Department of Periodontology, Academic Center for Dentistry Amsterdam (ACTA), University of Amsterdam and VU University, Amsterdam, the Netherlands
| |
Collapse
|
10
|
Chavassieux P, Chapurlat R. Interest of Bone Histomorphometry in Bone Pathophysiology Investigation: Foundation, Present, and Future. Front Endocrinol (Lausanne) 2022; 13:907914. [PMID: 35966102 PMCID: PMC9368205 DOI: 10.3389/fendo.2022.907914] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Accepted: 06/22/2022] [Indexed: 11/13/2022] Open
Abstract
Despite the development of non-invasive methods, bone histomorphometry remains the only method to analyze bone at the tissue and cell levels. Quantitative analysis of transiliac bone sections requires strict methodologic conditions but since its foundation more 60 years ago, this methodology has progressed. Our purpose was to review the evolution of bone histomorphometry over the years and its contribution to the knowledge of bone tissue metabolism under normal and pathological conditions and the understanding of the action mechanisms of therapeutic drugs in humans. The two main applications of bone histomorphometry are the diagnosis of bone diseases and research. It is warranted for the diagnosis of mineralization defects as in osteomalacia, of other causes of osteoporosis as bone mastocytosis, or the classification of renal osteodystrophy. Bone biopsies are required in clinical trials to evaluate the safety and mechanism of action of new therapeutic agents and were applied to anti-osteoporotic agents such as bisphosphonates and denosumab, an anti-RANKL, which induces a marked reduction of the bone turnover with a consequent elongation of the mineralization period. In contrast, an increased bone turnover with an extension of the formation site is observed with teriparatide. Romosozumab, an anti-sclerostin, has a dual effect with an early increased formation and reduced resorption. Bone histomorphometric studies allow us to understand the mechanism of coupling between formation and resorption and to evaluate the respective role of bone modeling and remodeling. The adaptation of new image analysis techniques will help bone biopsy analysis in the future.
Collapse
|
11
|
Lamarche BA, Thomsen JS, Andreasen CM, Lievers WB, Andersen TL. 2D size of trabecular bone structure units (BSU) correlate more strongly with 3D architectural parameters than age in human vertebrae. Bone 2022; 160:116399. [PMID: 35364343 DOI: 10.1016/j.bone.2022.116399] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 03/14/2022] [Accepted: 03/25/2022] [Indexed: 11/22/2022]
Abstract
Bone tissue is continuously remodeled. In trabecular bone, each remodeling transaction forms a microscopic bone structural unit (BSU), also known as a hemiosteon or a trabecular packet, which is bonded to existing tissue by osteopontin-rich cement lines. The size and shape of the BSUs are determined by the size and shape of the resorption cavity, and whether the cavity is potentially over- or under-filled by the subsequent bone formation. The present study focuses on the recently formed trabecular BSUs, and how their 2D size and shape changes with age and trabecular microstructure. The study was performed using osteopontin-immunostained frontal sections of L2 vertebrae from 8 young (aged 18.5-37.6 years) and 8 old (aged 69.1-96.4 years) control females, which underwent microcomputed tomography (μCT) imaging prior to sectioning. The contour of 4230 BSU profiles (181-385 per vertebra) within 1024 trabecular profiles were outlined, and their 2D width, length, area, and shape were assessed. Of these BSUs, 22 (0.5%) were generated by modeling-based bone formation (i.e. without prior resorption), while 99.5% were generated by remodeling-based bone formation (i.e. with prior resorption). The distributions of BSU profile width, length, and area were significantly smaller in the old versus young females (p < 0.005), and the median profile width, length, and area were negative correlated with age (p < 0.018). Importantly, these BSU profile size parameters were more strongly correlated with trabecular bone volume (BV/TV, p < 0.002) and structure model index (SMI, p < 0.008) assessed by μCT, than age. Moreover, the 2D BSU size parameters were positively correlated to the area of the individual trabecular profiles (p < 0.0001), which were significantly smaller in the old versus young females (p < 0.024). The BSU shape parameters (aspect ratio, circularity, and solidity) were not correlated with age, BV/TV, or SMI. Collectively, the study supports the notion that not only the BSU profile width, but also its length and area, are more influenced by the age-related bone loss and shift from plates to rods (SMI), than age itself. This implies that BSU profile size is mainly driven by changes in the trabecular microstructure, which affect the size of the resorption cavity that the BSU refills.
Collapse
Affiliation(s)
- Britney Alexi Lamarche
- Bharti School of Engineering and Computer Science, Laurentian University, Sudbury, Ontario, Canada
| | | | - Christina Møller Andreasen
- Clinical Cell Biology, Dept. of Pathology, Odense University Hospital, Odense, Denmark; Pathology Research Unit, Dept. of Molecular Medicine & Dept. of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - W Brent Lievers
- Bharti School of Engineering and Computer Science, Laurentian University, Sudbury, Ontario, Canada.
| | - Thomas Levin Andersen
- Clinical Cell Biology, Dept. of Pathology, Odense University Hospital, Odense, Denmark; Pathology Research Unit, Dept. of Molecular Medicine & Dept. of Clinical Research, University of Southern Denmark, Odense, Denmark; Dept. of Forensic Medicine, Aarhus University, Aarhus, Denmark.
| |
Collapse
|
12
|
Borggaard XG, Nielsen MH, Delaisse JM, Andreasen CM, Andersen TL. Spatial Organization of Osteoclastic Coupling Factors and Their Receptors at Human Bone Remodeling Sites. Front Mol Biosci 2022; 9:896841. [PMID: 35775083 PMCID: PMC9239410 DOI: 10.3389/fmolb.2022.896841] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Accepted: 05/11/2022] [Indexed: 11/17/2022] Open
Abstract
The strictly regulated bone remodeling process ensures that osteoblastic bone formation is coupled to osteoclastic bone resorption. This coupling is regulated by a panel of coupling factors, including clastokines promoting the recruitment, expansion, and differentiation of osteoprogenitor cells within the eroded cavity. The osteoprogenitor cells on eroded surfaces are called reversal cells. They are intermixed with osteoclasts and become bone-forming osteoblast when reaching a critical density and maturity. Several coupling factors have been proposed in the literature, but their effects and expression pattern vary between studies depending on species and experimental setup. In this study, we investigated the mRNA levels of proposed secreted and membrane-bound coupling factors and their receptors in cortical bone remodeling events within the femur of healthy adolescent human controls using high-sensitivity RNA in situ hybridization. Of the proposed coupling factors, human osteoclasts showed mRNA-presence of LIF, PDGFB, SEMA4D, but no presence of EFNB2, and OSM. On the other hand, the osteoblastic reversal cells proximate to osteoclasts presented with LIFR, PDGFRA and PLXNB1, but not PDGFRB, which are all known receptors of the proposed coupling factors. Although EFNB2 was not present in mature osteoclasts, the mRNA of the ligand-receptor pair EFNB2:EPHB4 were abundant near the central blood vessels within intracortical pores with active remodeling. EPHB4 and SEMA4D were also abundant in mature bone-forming osteoblasts. This study highlights that especially LIF:LIFR, PDGFB:PDGFRA, SEMA4D:PLXNB1 may play a critical role in the osteoclast-osteoblast coupling in human remodeling events, as they are expressed within the critical cells.
Collapse
Affiliation(s)
- Xenia G. Borggaard
- Research Unit of Pathology, Department of Clinical Research and Department of Molecular Medicine, Molecular Bone Histology Team, Clinical Cell Biology, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- *Correspondence: Xenia G. Borggaard, orcid.org/0000-0002-4922-2478 Thomas L. Andersen,
| | - Malene H. Nielsen
- Research Unit of Pathology, Department of Clinical Research and Department of Molecular Medicine, Molecular Bone Histology Team, Clinical Cell Biology, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Jean-Marie Delaisse
- Research Unit of Pathology, Department of Clinical Research and Department of Molecular Medicine, Molecular Bone Histology Team, Clinical Cell Biology, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Christina M. Andreasen
- Research Unit of Pathology, Department of Clinical Research and Department of Molecular Medicine, Molecular Bone Histology Team, Clinical Cell Biology, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Thomas L. Andersen
- Research Unit of Pathology, Department of Clinical Research and Department of Molecular Medicine, Molecular Bone Histology Team, Clinical Cell Biology, University of Southern Denmark, Odense, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Department of Forensic Medicine, Aarhus University, Aarhus, Denmark
- *Correspondence: Xenia G. Borggaard, orcid.org/0000-0002-4922-2478 Thomas L. Andersen,
| |
Collapse
|
13
|
Cheng CH, Chen LR, Chen KH. Osteoporosis Due to Hormone Imbalance: An Overview of the Effects of Estrogen Deficiency and Glucocorticoid Overuse on Bone Turnover. Int J Mol Sci 2022; 23:1376. [PMID: 35163300 PMCID: PMC8836058 DOI: 10.3390/ijms23031376] [Citation(s) in RCA: 230] [Impact Index Per Article: 76.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 01/14/2022] [Accepted: 01/24/2022] [Indexed: 02/07/2023] Open
Abstract
Osteoporosis is a serious health issue among aging postmenopausal women. The majority of postmenopausal women with osteoporosis have bone loss related to estrogen deficiency. The rapid bone loss results from an increase in bone turnover with an imbalance between bone resorption and bone formation. Osteoporosis can also result from excessive glucocorticoid usage, which induces bone demineralization with significant changes of spatial heterogeneities of bone at microscale, indicating potential risk of fracture. This review is a summary of current literature about the molecular mechanisms of actions, the risk factors, and treatment of estrogen deficiency related osteoporosis (EDOP) and glucocorticoid induced osteoporosis (GIOP). Estrogen binds with estrogen receptor to promote the expression of osteoprotegerin (OPG), and to suppress the action of nuclear factor-κβ ligand (RANKL), thus inhibiting osteoclast formation and bone resorptive activity. It can also activate Wnt/β-catenin signaling to increase osteogenesis, and upregulate BMP signaling to promote mesenchymal stem cell differentiation from pre-osteoblasts to osteoblasts, rather than adipocytes. The lack of estrogen will alter the expression of estrogen target genes, increasing the secretion of IL-1, IL-6, and tumor necrosis factor (TNF). On the other hand, excessive glucocorticoids interfere the canonical BMP pathway and inhibit Wnt protein production, causing mesenchymal progenitor cells to differentiate toward adipocytes rather than osteoblasts. It can also increase RANKL/OPG ratio to promote bone resorption by enhancing the maturation and activation of osteoclast. Moreover, excess glucocorticoids are associated with osteoblast and osteocyte apoptosis, resulting in declined bone formation. The main focuses of treatment for EDOP and GIOP are somewhat different. Avoiding excessive glucocorticoid use is mandatory in patients with GIOP. In contrast, appropriate estrogen supplement is deemed the primary treatment for females with EDOP of various causes. Other pharmacological treatments include bisphosphonate, teriparatide, and RANKL inhibitors. Nevertheless, more detailed actions of EDOP and GIOP along with the safety and effectiveness of medications for treating osteoporosis warrant further investigation.
Collapse
Affiliation(s)
- Chu-Han Cheng
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan; (C.-H.C.); (L.-R.C.)
| | - Li-Ru Chen
- Department of Physical Medicine and Rehabilitation, Mackay Memorial Hospital, Taipei 104, Taiwan; (C.-H.C.); (L.-R.C.)
- Department of Mechanical Engineering, National Yang Ming Chiao Tung University, Hsinchu 300, Taiwan
| | - Kuo-Hu Chen
- Department of Obstetrics and Gynecology, Taipei Tzu-Chi Hospital, The Buddhist Tzu-Chi Medical Foundation, Taipei 231, Taiwan
- School of Medicine, Tzu-Chi University, Hualien 970, Taiwan
| |
Collapse
|
14
|
Sun Y, Li J, Xie X, Gu F, Sui Z, Zhang K, Yu T. Recent Advances in Osteoclast Biological Behavior. Front Cell Dev Biol 2021; 9:788680. [PMID: 34957116 PMCID: PMC8694526 DOI: 10.3389/fcell.2021.788680] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Accepted: 11/15/2021] [Indexed: 12/19/2022] Open
Abstract
With the progress of the aging population, bone-related diseases such as osteoporosis and osteoarthritis have become urgent problems. Recent studies have demonstrated the importance of osteoclasts in bone homeostasis, implying these will be an important mediator in the treatment of bone-related diseases. Up to now, several reviews have been performed on part of osteoclast biological behaviors such as differentiation, function, or apoptosis. However, few reviews have shown the complete osteoclast biology and research advances in recent years. Therefore, in this review, we focus on the origin, differentiation, apoptosis, behavior changes and coupling signals with osteoblasts, providing a simple but comprehensive overview of osteoclasts for subsequent studies.
Collapse
Affiliation(s)
- Yang Sun
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Jiangbi Li
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Xiaoping Xie
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Feng Gu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Zhenjiang Sui
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Ke Zhang
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| | - Tiecheng Yu
- Department of Orthopedics, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
15
|
Povoroznyuk VV, Dedukh NV, Bystrytska MA, Shapovalov VS. Bone remodeling stages under physiological conditions and glucocorticoid in excess: Focus on cellular and molecular mechanisms. REGULATORY MECHANISMS IN BIOSYSTEMS 2021. [DOI: 10.15421/022130] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
This review provides a rationale for the cellular and molecular mechanisms of bone remodeling stages under physiological conditions and glucocorticoids (GCs) in excess. Remodeling is a synchronous process involving bone resorption and formation, proceeding through stages of: (1) resting bone, (2) activation, (3) bone resorption, (4) reversal, (5) formation, (6) termination. Bone remodeling is strictly controlled by local and systemic regulatory signaling molecules. This review presents current data on the interaction of osteoclasts, osteoblasts and osteocytes in bone remodeling and defines the role of osteoprogenitor cells located above the resorption area in the form of canopies and populating resorption cavities. The signaling pathways of proliferation, differentiation, viability, and cell death during remodeling are presented. The study of signaling pathways is critical to understanding bone remodeling under normal and pathological conditions. The main signaling pathways that control bone resorption and formation are RANK / RANKL / OPG; M-CSF – c-FMS; canonical and non-canonical signaling pathways Wnt; Notch; MARK; TGFβ / SMAD; ephrinB1/ephrinB2 – EphB4, TNFα – TNFβ, and Bim – Bax/Bak. Cytokines, growth factors, prostaglandins, parathyroid hormone, vitamin D, calcitonin, and estrogens also act as regulators of bone remodeling. The role of non-encoding microRNAs and long RNAs in the process of bone cell differentiation has been established. MicroRNAs affect many target genes, have both a repressive effect on bone formation and activate osteoblast differentiation in different ways. Excess of glucocorticoids negatively affects all stages of bone remodeling, disrupts molecular signaling, induces apoptosis of osteocytes and osteoblasts in different ways, and increases the life cycle of osteoclasts. Glucocorticoids disrupt the reversal stage, which is critical for the subsequent stages of remodeling. Negative effects of GCs on signaling molecules of the canonical Wingless (WNT)/β-catenin pathway and other signaling pathways impair osteoblastogenesis. Under the influence of excess glucocorticoids biosynthesis of biologically active growth factors is reduced, which leads to a decrease in the expression by osteoblasts of molecules that form the osteoid. Glucocorticoids stimulate the expression of mineralization inhibitor proteins, osteoid mineralization is delayed, which is accompanied by increased local matrix demineralization. Although many signaling pathways involved in bone resorption and formation have been discovered and described, the temporal and spatial mechanisms of their sequential turn-on and turn-off in cell proliferation and differentiation require additional research.
Collapse
|
16
|
Jensen PR, Andersen TL, Chavassieux P, Roux JP, Delaisse JM. Bisphosphonates impair the onset of bone formation at remodeling sites. Bone 2021; 145:115850. [PMID: 33465485 DOI: 10.1016/j.bone.2021.115850] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 01/08/2021] [Accepted: 01/15/2021] [Indexed: 12/20/2022]
Abstract
Bisphosphonates are widely used anti-osteoporotic drugs targeting osteoclasts. They strongly inhibit bone resorption, but also strongly reduce bone formation. This reduced formation is commonly ascribed to the mechanism maintaining the resorption/formation balance during remodeling. The present study provides evidence for an additional mechanism where bisphosphonates actually impair the onset of bone formation after resorption. The evidence is based on morphometric parameters recently developed to assess the activities reversing resorption to formation. Herein, we compare these parameters in cancellous bone of alendronate- and placebo-treated postmenopausal osteoporotic patients. Alendronate increases the prevalence of eroded surfaces characterized by reversal cells/osteoprogenitors at low cell density and remote from active bone surfaces. This indicates deficient cell expansion on eroded surfaces - an event that is indispensable to start formation. Furthermore, alendronate decreases the coverage of these eroded surfaces by remodeling compartment canopies, a putative source of reversal cells/osteoprogenitors. Finally, alendronate strongly decreases the activation frequency of bone formation, and decreases more the formative compared to the eroded surfaces. All these parameters correlate with each other. These observations lead to a model where bisphosphonates hamper the osteoprogenitor recruitment required to initiate bone formation. This effect results in a larger eroded surface, thereby explaining the well-known paradox that bisphosphonates strongly inhibit bone resorption without strongly decreasing eroded surfaces. The possible mechanism for hampered osteoprogenitor recruitment is discussed: bisphosphonates may decrease the release of osteogenic factors by the osteoclasts, and/or bisphosphonates released by osteoclasts may act directly on neighboring osteoprogenitor cells as reported in preclinical studies.
Collapse
Affiliation(s)
- Pia Rosgaard Jensen
- Clinical Cell Biology, Lillebælt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark.
| | - Thomas Levin Andersen
- Clinical Cell Biology, Lillebælt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark; Clinical Cell Biology, Department of Pathology, Odense University Hospital, Department of Clinical Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Forensic Medicine, Aarhus University, Aarhus, Denmark.
| | | | | | - Jean-Marie Delaisse
- Clinical Cell Biology, Lillebælt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark; Clinical Cell Biology, Department of Pathology, Odense University Hospital, Department of Clinical Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
17
|
Delaisse JM, Andersen TL, Kristensen HB, Jensen PR, Andreasen CM, Søe K. Re-thinking the bone remodeling cycle mechanism and the origin of bone loss. Bone 2020; 141:115628. [PMID: 32919109 DOI: 10.1016/j.bone.2020.115628] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 09/04/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
Abstract
Proper bone remodeling necessarily requires that osteoblasts reconstruct the bone that osteoclasts have resorbed. However, the cellular events connecting resorption to reconstruction have remained poorly known. The consequence is a fragmentary understanding of the remodeling cycle where only the resorption and formation steps are taken into account. New tools have recently made possible to elucidate how resorption shifts to formation, thereby allowing to comprehend the remodeling cycle as a whole. This new knowledge is reviewed herein. It shows how teams of osteoclasts and osteoblast lineage cells are progressively established and how they are subjected therein to reciprocal interactions. Contrary to the common view, osteoclasts and osteoprogenitors are intermingled on the eroded surfaces. The analysis of the resorption and cell population dynamics shows that osteoprogenitor cell expansion and resorption proceed as an integrated mechanism; that a threshold cell density of osteoprogenitors on the eroded surface is mandatory for onset of bone formation; that the cell initiating osteoprogenitor cell expansion is the osteoclast; and that the osteoclast therefore triggers putative osteoprogenitor reservoirs positioned at proximity of the eroded bone surface (bone lining cells, canopy cells, pericytes). The interplay between magnitude of resorption and rate of cell expansion governs how soon bone reconstruction is initiated and may determine uncoupling and permanent bone loss if a threshold cell density is not reached. The clinical perspectives opened by these findings are discussed.
Collapse
Affiliation(s)
- Jean-Marie Delaisse
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Department of Clinical Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Thomas Levin Andersen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Department of Clinical Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark; Department of Forensic Medicine, Aarhus University, Aarhus, Denmark.
| | - Helene Bjoerg Kristensen
- Clinical Cell Biology, Lillebælt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark.
| | - Pia Rosgaard Jensen
- Clinical Cell Biology, Lillebælt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark.
| | - Christina Møller Andreasen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Department of Clinical Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Department of Clinical Research, Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
18
|
Harrison KD, Hiebert BD, Panahifar A, Andronowski JM, Ashique AM, King GA, Arnason T, Swekla KJ, Pivonka P, Cooper DM. Cortical Bone Porosity in Rabbit Models of Osteoporosis. J Bone Miner Res 2020; 35:2211-2228. [PMID: 32614975 PMCID: PMC7702175 DOI: 10.1002/jbmr.4124] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Revised: 06/17/2020] [Accepted: 06/21/2020] [Indexed: 12/20/2022]
Abstract
Cortical bone porosity is intimately linked with remodeling, is of growing clinical interest, and is increasingly accessible by imaging. Thus, the potential of animal models of osteoporosis (OP) to provide a platform for studying how porosity develops and responds to interventions is tremendous. To date, rabbit models of OP have largely focused on trabecular microarchitecture or bone density; some such as ovariectomy (OVX) have uncertain efficacy and cortical porosity has not been extensively reported. Our primary objective was to characterize tibial cortical porosity in rabbit-based models of OP, including OVX, glucocorticoids (GC), and OVX + GC relative to controls (SHAM). We sought to: (i) test the hypothesis that intracortical remodeling is elevated in these models; (ii) contrast cortical remodeling and porosity in these models with that induced by parathyroid hormone (1-34; PTH); and (iii) contrast trabecular morphology in the proximal tibia across all groups. Evidence that an increase in cortical porosity occurred in all groups was observed, although this was the least robust for GC. Histomorphometric measures supported the hypothesis that remodeling rate was elevated in all groups and also revealed evidence of uncoupling of bone resorption and formation in the GC and OVX + GC groups. For trabecular bone, a pattern of loss was observed for OVX, GC, and OVX + GC groups, whereas the opposite was observed for PTH. Change in trabecular number best explained these patterns. Taken together, the findings indicated rabbit models provide a viable and varied platform for the study of OP and associated changes in cortical remodeling and porosity. Intriguingly, the evidence revealed differing effects on the cortical and trabecular envelopes for the PTH model. © 2020 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR)..
Collapse
Affiliation(s)
- Kim D Harrison
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Beverly D Hiebert
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Arash Panahifar
- BioMedical Imaging and Therapy Beamline, Canadian Light Source, Saskatoon, Canada.,Department of Medical Imaging, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | | | | | - Gavin A King
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Terra Arnason
- Department of Medicine, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| | - Kurtis J Swekla
- Research Services and Ethics Office, Office of the Vice President of Research, University of Saskatchewan, Saskatoon, Canada
| | - Peter Pivonka
- School of Mechanical, Medical, and Process Engineering, Queensland University of Technology, Brisbane, Australia
| | - David Ml Cooper
- Department of Anatomy, Physiology, and Pharmacology, College of Medicine, University of Saskatchewan, Saskatoon, Canada
| |
Collapse
|
19
|
Fassio A, Adami G, Idolazzi L, Giollo A, Viapiana O, Vantaggiato E, Benini C, Rossini M, Dejaco C, Gatti D. Wnt Inhibitors and Bone Turnover Markers in Patients With Polymyalgia Rheumatica and Acute Effects of Glucocorticoid Treatment. Front Med (Lausanne) 2020; 7:551. [PMID: 33015101 PMCID: PMC7509037 DOI: 10.3389/fmed.2020.00551] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 07/31/2020] [Indexed: 12/12/2022] Open
Abstract
Background: In polymyalgia rheumatica (PMR), data on bone turnover markers (BTM), on Wnt inhibitors (Dkk-1, sclerostin) and their changes induced by glucocorticoids (GC) are lacking. The aims of our study were to compare the baseline levels of Wnt inhibitors and BTM in PMR patients with healthy controls (HC) and to study their changes over the first 4 weeks of GC treatment. Materials and Methods: We enrolled 17 treatment-naïve patients affected by PMR and 17 age and sex-matched healthy controls (HC) from retired hospital personnel. PMR patients were administered methylprednisolone 16 mg daily for 4 weeks. Blood samples were taken at baseline and at week 4 for the PMR group, a single sample was taken for HC. N-propeptide of type I collagen (PINP), C-terminal telopeptide of type I collagen (CTX-I), sclerostin, Dkk-1, and C-reactive protein (CRP) were dosed. Results: At baseline, Dkk-1 was significantly higher in the PMR group as compared to HC (p = 0.002) while PINP, CTX-I and sclerostin levels were comparable between PMR patients and HC, After 4 weeks of GC treatment we found in the PMR group a decrease of PINP (mean ± SD percentage decrement as compared to baseline −40 ± 18.6%, p < 0.001), CTX-I (−23.5 ± 41.3%, p = 0.032), Dkk-1 (−22.4 ± 39.6, p = 0.033), and sclerostin (−32.49 ± 20.47, p < 0.001) as compared to baseline levels. Conclusions: In treatment-naïve PMR, systemic inflammation is associated with a dysregulation of the Wnt system (increased Dkk-1). Within the 1st month, treatment with GC showed noteworthy effects on bone resorption, formation, and on Wnt pathway modulators.
Collapse
Affiliation(s)
- Angelo Fassio
- Rheumatology Unit, University of Verona, Verona, Italy
| | | | - Luca Idolazzi
- Rheumatology Unit, University of Verona, Verona, Italy
| | | | | | | | | | | | - Christian Dejaco
- Rheumatology Service, South Tyrolean Health Trust, Hospital of Bruneck, Bruneck, Italy.,Department of Rheumatology, Medical University Graz, Graz, Austria
| | - Davide Gatti
- Rheumatology Unit, University of Verona, Verona, Italy
| |
Collapse
|
20
|
Borciani G, Montalbano G, Baldini N, Cerqueni G, Vitale-Brovarone C, Ciapetti G. Co-culture systems of osteoblasts and osteoclasts: Simulating in vitro bone remodeling in regenerative approaches. Acta Biomater 2020; 108:22-45. [PMID: 32251782 DOI: 10.1016/j.actbio.2020.03.043] [Citation(s) in RCA: 122] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 03/20/2020] [Accepted: 03/30/2020] [Indexed: 02/08/2023]
Abstract
Bone is an extremely dynamic tissue, undergoing continuous remodeling for its whole lifetime, but its regeneration or augmentation due to bone loss or defects are not always easy to obtain. Bone tissue engineering (BTE) is a promising approach, and its success often relies on a "smart" scaffold, as a support to host and guide bone formation through bone cell precursors. Bone homeostasis is maintained by osteoblasts (OBs) and osteoclasts (OCs) within the basic multicellular unit, in a consecutive cycle of resorption and formation. Therefore, a functional scaffold should allow the best possible OB/OC cooperation for bone remodeling, as happens within the bone extracellular matrix in the body. In the present work OB/OC co-culture models, with and without scaffolds, are reviewed. These experimental systems are intended for different targets, including bone remodeling simulation, drug testing and the assessment of biomaterials and 3D scaffolds for BTE. As a consequence, several parameters, such as cell type, cell ratio, culture medium and inducers, culture times and setpoints, assay methods, etc. vary greatly. This review identifies and systematically reports the in vitro methods explored up to now, which, as they allow cellular communication, more closely resemble bone remodeling and/or the regeneration process in the framework of BTE. STATEMENT OF SIGNIFICANCE: Bone is a dynamic tissue under continuous remodeling, but spontaneous healing may fail in the case of excessive bone loss which often requires valid alternatives to conventional treatments to restore bone integrity, like bone tissue engineering (BTE). Pre-clinical evaluation of scaffolds for BTE requires in vitro testing where co-cultures combining innovative materials with osteoblasts (OBs) and osteoclasts (OCs) closely mimic the in vivo repair process. This review considers the direct and indirect OB/OC co-cultures relevant to BTE, from the early mouse-cell models to the recent bone regenerative systems. The co-culture modeling of bone microenvironment provides reliable information on bone cell cross-talk. Starting from improved knowledge on bone remodeling, bone disease mechanisms may be understood and new BTE solutions are designed.
Collapse
Affiliation(s)
- Giorgia Borciani
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Giorgia Montalbano
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy
| | - Nicola Baldini
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Italy
| | - Giorgia Cerqueni
- Department of Clinical and Molecular Sciences (DISCLIMO), Università Politecnica delle Marche, Via Tronto 10/a, Ancona 60020, Italy
| | - Chiara Vitale-Brovarone
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, Torino 10129, Italy.
| | - Gabriela Ciapetti
- Laboratory for Orthopaedic Pathophysiology and Regenerative Medicine, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| |
Collapse
|
21
|
Conaway HH, Henning P, Lie A, Tuckermann J, Lerner UH. Glucocorticoids employ the monomeric glucocorticoid receptor to potentiate vitamin D 3 and parathyroid hormone-induced osteoclastogenesis. FASEB J 2019; 33:14394-14409. [PMID: 31675485 PMCID: PMC6894088 DOI: 10.1096/fj.201802729rrr] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Glucocorticoid (GC) therapy decreases bone mass and increases the risk of fractures. We investigated interactions between the GC dexamethasone (DEX) and the bone resorptive agents 1,25(OH)2-vitamin D3 (D3) and parathyroid hormone (PTH) on osteoclastogenesis. We observed a synergistic potentiation of osteoclast progenitor cell differentiation and formation of osteoclasts when DEX was added to either D3- or PTH-treated mouse bone marrow cell (BMC) cultures. Cotreatment of DEX with D3 or PTH increased gene encoding calcitonin receptor (Calcr), acid phosphatase 5, tartrate resistant (Acp5), cathepsin K (Ctsk), and TNF superfamily member 11 (Tnfsf11) mRNA, receptor activator of NF-κB ligand protein (RANKL), numbers of osteoclasts on plastic, and pit formation and release of C-terminal fragment of type I collagen from cells cultured on bone slices. Enhanced RANKL protein expression caused by D3 and DEX was absent in BMC from mice in which the GC receptor (GR) was deleted in stromal cells/osteoblasts. Synergistic interactions between DEX and D3 on RANKL and osteoclast formation were present in BMC from mice with attenuated GR dimerization. These data demonstrate that the GR cooperates with D3 and PTH signaling, causing massive osteoclastogenesis, which may explain the rapid bone loss observed with high dosages of GC treatment.-Conaway, H. H., Henning, P., Lie, A., Tuckermann, J., Lerner, U. H. Glucocorticoids employ the monomeric glucocorticoid receptor to potentiate vitamin D3 and parathyroid hormone-induced osteoclastogenesis.
Collapse
Affiliation(s)
- H Herschel Conaway
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Petra Henning
- Center for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Antia Lie
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Ulf H Lerner
- Center for Bone and Arthritis Research, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden.,Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| |
Collapse
|
22
|
Sims NA, Martin TJ. Osteoclasts Provide Coupling Signals to Osteoblast Lineage Cells Through Multiple Mechanisms. Annu Rev Physiol 2019; 82:507-529. [PMID: 31553686 DOI: 10.1146/annurev-physiol-021119-034425] [Citation(s) in RCA: 173] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bone remodeling is essential for the repair and replacement of damaged and old bone. The major principle underlying this process is that osteoclast-mediated resorption of a quantum of bone is followed by osteoblast precursor recruitment; these cells differentiate to matrix-producing osteoblasts, which form new bone to replace what was resorbed. Evidence from osteopetrotic syndromes indicate that osteoclasts not only resorb bone, but also provide signals to promote bone formation. Osteoclasts act upon osteoblast lineage cells throughout their differentiation by facilitating growth factor release from resorbed matrix, producing secreted proteins and microvesicles, and expressing membrane-bound factors. These multiple mechanisms mediate the coupling of bone formation to resorption in remodeling. Additional interactions of osteoclasts with osteoblast lineage cells, including interactions with canopy and reversal cells, are required to achieve coordination between bone formation and resorption during bone remodeling.
Collapse
Affiliation(s)
- Natalie A Sims
- Bone Cell Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; , .,Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia
| | - T John Martin
- Bone Cell Biology and Disease Unit, St. Vincent's Institute of Medical Research, Melbourne, Victoria 3065, Australia; , .,Department of Medicine, The University of Melbourne, St. Vincent's Hospital, Melbourne, Victoria 3065, Australia
| |
Collapse
|
23
|
Frost M, Tencerova M, Andreasen CM, Andersen TL, Ejersted C, Svaneby D, Qui W, Kassem M, Zarei A, McAlister WH, Veis DJ, Whyte MP, Frederiksen AL. Absence of an osteopetrosis phenotype in IKBKG (NEMO) mutation-positive women: A case-control study. Bone 2019; 121:243-254. [PMID: 30659980 PMCID: PMC6457251 DOI: 10.1016/j.bone.2019.01.014] [Citation(s) in RCA: 3] [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: 08/23/2018] [Revised: 12/12/2018] [Accepted: 01/14/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND NF-κB essential modulator (NEMO), encoded by IKBKG, is necessary for activation of the ubiquitous transcription factor nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). Animal studies suggest NEMO is required for NF-κB mediated bone homeostasis, but this has not been thoroughly studied in humans. IKBKG loss-of-function mutation causes incontinentia pigmenti (IP), a rare X-linked disease featuring linear hypopigmentation, alopecia, hypodontia, and immunodeficiency. Single case reports describe osteopetrosis (OPT) in boys carrying hypomorphic IKBKG mutations. METHOD We studied the bone phenotype in women with IP with evaluation of radiographs of the spine and non-dominant arm and leg; lumbar spine and femoral neck aBMD using DXA; μ-CT and histomorphometry of trans-iliac crest biopsy specimens; bone turnover markers; and cellular phenotype in bone marrow skeletal (stromal) stem cells (BM-MSCs) in a cross-sectional, age-, sex-, and BMI-matched case-control study. X-chromosome inactivation was measured in blood leucocytes and BM-MSCs using a PCR method with methylation of HpaII sites. NF-κB activity was quantitated in BM-MSCs using a luciferase NF-κB reporter assay. RESULTS Seven Caucasian women with IP (age: 24-67 years and BMI: 20.0-35.2 kg/m2) and IKBKG mutation (del exon 4-10 (n = 4); c.460C>T (n = 3)) were compared to matched controls. The IKBKG mutation carriers had extremely skewed X-inactivation (>90:10%) in blood, but not in BM-MSCs. NF-κB activity was lower in BM-MSCs from IKBKG mutation carriers (n = 5) compared to controls (3094 ± 679 vs. 5422 ± 1038/μg protein, p < 0.01). However, no differences were identified on skeletal radiographics, aBMD, μ-architecture of the iliac crest, or bone turnover markers. The IKBKG mutation carriers had a 1.7-fold greater extent of eroded surfaces relative to osteoid surfaces (p < 0.01), and a 2.0-fold greater proportion of arrested reversal surface relative to active reversal surface (p < 0.01). CONCLUSION Unlike mutation-positive males, the IKBKG mutation-positive women did not manifest OPT.
Collapse
Affiliation(s)
- Morten Frost
- Department of Clinical Research, Faculty of Health, University of Southern Denmark (SDU), Winsløwparken 19. 3, DK-5000 Odense C, Denmark; Steno Diabetes Center Odense, Odense University Hospital (OUH), J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark; Department of Endocrinology, Molecular Endocrinology Unit, OUH, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark.
| | - Michaela Tencerova
- Department of Endocrinology, Molecular Endocrinology Unit, OUH, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark.
| | - Christina M Andreasen
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery & Traumatology, OUH, J.B. Winsløws Vej 15, DK-5000 Odense C, Denmark; Department of Clinical Cell Biology, Vejle Hospital, Beridderbakken 4, DK-7100 Vejle, Denmark.
| | - Thomas L Andersen
- Department of Clinical Cell Biology, Vejle Hospital, Beridderbakken 4, DK-7100 Vejle, Denmark.
| | - Charlotte Ejersted
- Department of Endocrinology, Molecular Endocrinology Unit, OUH, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark.
| | - Dea Svaneby
- Department of Clinical Genetics, Vejle Hospital, Beridderbakken 4, DK-7100 Vejle, Denmark.
| | - Weimin Qui
- Department of Endocrinology, Molecular Endocrinology Unit, OUH, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark
| | - Moustapha Kassem
- Department of Endocrinology, Molecular Endocrinology Unit, OUH, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark.
| | - Allahdad Zarei
- Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, MO, USA.
| | - William H McAlister
- Department of Pediatric Radiology, Mallinckrodt Institute of Radiology, Washington University School of Medicine at St. Louis Children's Hospital, St. Louis, MO, USA.
| | - Deborah J Veis
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, St. Louis, MO, USA; Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, MO, USA.
| | - Michael P Whyte
- Center for Metabolic Bone Disease and Molecular Research, Shriners Hospital for Children, St. Louis, MO, USA; Division of Bone and Mineral Diseases, Department of Internal Medicine, Washington University School of Medicine at Barnes-Jewish Hospital, St. Louis, MO, USA.
| | - Anja L Frederiksen
- Department of Clinical Research, Faculty of Health, University of Southern Denmark (SDU), Winsløwparken 19. 3, DK-5000 Odense C, Denmark; Department of Clinical Genetics, Odense University Hospital, J.B. Winsløws Vej 4, DK-5000 Odense C, Denmark.
| |
Collapse
|
24
|
Bakalova LP, Andreasen CM, Thomsen JS, Brüel A, Hauge EM, Kiil BJ, Delaisse JM, Andersen TL, Kersh ME. Intracortical Bone Mechanics Are Related to Pore Morphology and Remodeling in Human Bone. J Bone Miner Res 2018; 33:2177-2185. [PMID: 30048570 DOI: 10.1002/jbmr.3561] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/11/2018] [Accepted: 07/20/2018] [Indexed: 01/06/2023]
Abstract
During aging and in osteoporosis, cortical bone becomes more porous, making it more fragile and susceptible to fractures. The aim of this study was to investigate the intracortical compression- induced strain energy distribution, and determine whether intracortical pores associated with high strain energy density (SED) in the surrounding bone matrix have a different morphology and distribution, as well as different remodeling characteristics than matrix with normal SED. Fibular diaphyseal specimens from 20 patients undergoing a jaw reconstruction (age range 41 to 75 years; 14 men and 6 women) were studied. Bone specimens were µCT-scanned, plastic embedded, and sectioned for histology. Three-dimensional microfinite element models of each specimen were tested in compression, and the SED of the bone immediately surrounding the intracortical pores was calculated within a plane of interest corresponding to the histological sections. The SED of a pore, relative to the distribution of the SED of all pores in each specimen, was used to classify pores as either a high or normal SED pore. Pores with high SED were larger, less circular, and were located closer to the endosteal surface of the cortex than normal SED pores (p < 0.001). Histological analysis of the remodeling events generating the pores revealed that the high SED pores compared with normal SED pores had 13.3-fold higher odds of being an erosive (70%) or formative (7%) pore versus a quiescent pore (p < 0.001), 5.9-fold higher odds of resulting from remodeling upon existing pores (type 2 pore) versus remodeling generating new pores (type 1 pore) (p < 0.001), and 3.2-fold higher odds of being a coalescing type 2 pore versus a noncoalescing type 2 pore (p < 0.001). Overall, the study demonstrates a strong relationship between cortical bone mechanics and pore morphology, distribution, and remodeling characteristics in human fibular bone. © 2018 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Lydia P Bakalova
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| | - Christina M Andreasen
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Institute of Clinical Research, University of Southern Denmark, Denmark
| | | | - Annemarie Brüel
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Ellen-Margrethe Hauge
- Department of Rheumatology, Aarhus University Hospital, and Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Birgitte Jul Kiil
- Department of Plastic Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Jean-Marie Delaisse
- Department of Clinical Cell Biology (KCB), Vejle Hospital-Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology (KCB), Vejle Hospital-Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Mariana E Kersh
- Department of Mechanical Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, USA.,Beckman Institute of Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, USA
| |
Collapse
|
25
|
Andreasen CM, Delaisse JM, van der Eerden BCJ, van Leeuwen JPTM, Ding M, Andersen TL. Understanding age-induced cortical porosity in women: Is a negative BMU balance in quiescent osteons a major contributor? Bone 2018; 117:70-82. [PMID: 30240959 DOI: 10.1016/j.bone.2018.09.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 09/10/2018] [Accepted: 09/16/2018] [Indexed: 01/18/2023]
Abstract
Cortical bone is remodeled by intracortical basic multicellular units (BMUs), whose end result can be observed as quiescent osteons in histological sections. These osteons offer a unique opportunity to investigate the BMU balance between the magnitude of bone resorption and subsequent bone formation at the BMU level. Our main objective was to investigate whether the latter parameters change between defined categories of osteons and with age, and to which extend these changes contribute to age-induced cortical porosity. Cortices of iliac bone specimens from 35 women (aged 16-78 years) with a higher porosity with age were investigated. A total of 3084 quiescent osteons reflecting 75% of the intracortical pores were histological examined. The osteons diameter, pore diameter, wall thickness, prevalence and contribution to the porosity were highly variable, but unchanged with age. Next, the osteons were categorized according to whether they reflected the remodeling of existing canals (type 2Q osteons) or the generation of new canals (type 1Q osteons). Type 2Q osteons versus type 1Q osteons: (i) had more frequently a pore diameter > 75 μm (7.4 vs. 1.3%; p < 0.001); (ii) had a larger mean pore diameter (40 ± 10 vs. 25 ± 4 μm; p < 0.001), osteon diameter (120 ± 21 vs. 94 ± 21 μm; p < 0.001) and wall thickness (40 ± 10 vs. 35 ± 9; p < 0.05); (iii) had a larger contribution to the cortical porosity (29 ± 18 vs. 8 ± 8%; p < 0.001); (iv) were more prevalent (44 ± 10 vs. 31 ± 11%; p < 0.001); and (v) were more prevalent with age. Collectively, this study demonstrates that quiescent osteons with age more frequently result from remodeling of existing canals, which in some cases had a more negative BMU balance. Still, the osteons showed no overall age-related change in their pore diameter i.e. BMU balance. In contrast to conventional wisdom, these data show that non-quiescent pores, not pores of quiescent osteons, were the main contributor to a higher cortical porosity.
Collapse
Affiliation(s)
- Christina M Andreasen
- Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark; Orthopaedic Research Laboratory, Department of Orthopaedic Surgery & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Denmark.
| | - Jean-Marie Delaisse
- Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark.
| | - Bram C J van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands.
| | - Johannes P T M van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus MC, Rotterdam, the Netherlands.
| | - Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Denmark.
| | - Thomas L Andersen
- Clinical Cell Biology, Vejle Hospital - Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, Vejle, Denmark.
| |
Collapse
|
26
|
Ohlsson C, Nilsson KH, Henning P, Wu J, Gustafsson KL, Poutanen M, Lerner UH, Movérare-Skrtic S. WNT16 overexpression partly protects against glucocorticoid-induced bone loss. Am J Physiol Endocrinol Metab 2018; 314:E597-E604. [PMID: 29406783 DOI: 10.1152/ajpendo.00292.2017] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Therapeutic use of glucocorticoids (GCs) is a major cause of secondary osteoporosis, but the molecular mechanisms responsible for the deleterious effects of GCs in bone are only partially understood. WNT16 is a crucial physiological regulator of bone mass and fracture susceptibility, and we hypothesize that disturbed WNT16 activity might be involved in the deleterious effects of GC in bone. Twelve-week-old female Obl-Wnt16 mice (WNT16 expression driven by the rat procollagen type I α1 promoter) and wild-type (WT) littermates were treated with prednisolone (7.6 mg·kg-1·day-1) or vehicle for 4 wk. We first observed that GC treatment decreased the Wnt16 mRNA levels in bone of female mice (-56.4 ± 6.1% compared with vehicle, P < 0.001). We next evaluated if WNT16 overexpression protects against GC-induced bone loss. Dual-energy X-ray absorptiometry analyses revealed that GC treatment decreased total body bone mineral density in WT mice (-3.9 ± 1.2%, P = 0.028) but not in Obl-Wnt16 mice (+1.3 ± 1.4%, nonsignificant). Microcomputed tomography analyses showed that GC treatment decreased trabecular bone volume fraction (BV/TV) of the femur in WT mice ( P = 0.019) but not in Obl-Wnt16 mice. Serum levels of the bone formation marker procollagen type I N-terminal propeptide were substantially reduced by GC treatment in WT mice (-50.3 ± 7.0%, P = 0.008) but not in Obl-Wnt16 mice (-3.8 ± 21.2%, nonsignificant). However, the cortical bone thickness in femur was reduced by GC treatment in both WT mice and Obl-Wnt16 mice. In conclusion, GC treatment decreases Wnt16 mRNA levels in bone and WNT16 overexpression partly protects against GC-induced bone loss.
Collapse
Affiliation(s)
- Claes Ohlsson
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Karin H Nilsson
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Petra Henning
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Jianyao Wu
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Karin L Gustafsson
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Matti Poutanen
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
- Institute of Biomedicine and Turku Center for Disease Modeling, University of Turku , Turku , Finland
| | - Ulf H Lerner
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| | - Sofia Movérare-Skrtic
- Centre for Bone and Arthritis Research at Institute of Medicine, Sahlgrenska Academy at University of Gothenburg , Gothenburg , Sweden
| |
Collapse
|
27
|
Xiang L, Yu H, Zhang X, Wang B, Yuan Y, Zhang Q, Ye R, Gong P, Wu Y. The versatile hippo pathway in oral-maxillofacial development and bone remodeling. Dev Biol 2018; 440:53-63. [PMID: 29792855 DOI: 10.1016/j.ydbio.2018.05.017] [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: 03/15/2018] [Revised: 05/20/2018] [Accepted: 05/20/2018] [Indexed: 02/05/2023]
Abstract
The Hippo signaling pathway is implicated in key aspects of cell proliferation, control of organ size, stem cell functions and tumor suppression. Its functions are primarily mediated either through direct effects on transcription factors to influence target gene expression or through crosstalk with other signaling pathways that regulate multiple physiological activities. Studies are revealing Hippo pathway involvement in diverse functions including renewal of intestinal epithelium, promotion of myocardial cell proliferation, cancer suppression, etc. In this review we discuss Hippo pathway signaling in oral-maxillofacial development and bone remodeling under normal and pathological conditions and highlight promising future research directions.
Collapse
Affiliation(s)
- Lin Xiang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Hui Yu
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xinyuan Zhang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Bin Wang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ying Yuan
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qin Zhang
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Rui Ye
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Ping Gong
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| | - Yingying Wu
- State Key Laboratory of Oral Diseases&National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
| |
Collapse
|
28
|
Andreasen CM, Delaisse JM, van der Eerden BC, van Leeuwen JP, Ding M, Andersen TL. Understanding Age-Induced Cortical Porosity in Women: The Accumulation and Coalescence of Eroded Cavities Upon Existing Intracortical Canals Is the Main Contributor. J Bone Miner Res 2018; 33:606-620. [PMID: 29193312 DOI: 10.1002/jbmr.3354] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Revised: 11/17/2017] [Accepted: 11/27/2017] [Indexed: 01/08/2023]
Abstract
Intracortical bone remodeling normally ensures maintenance of the cortical bone matrix and strength, but during aging, this remodeling generates excessive porosity. The mechanism behind the age-induced cortical porosity is poorly understood and addressed in the present study. This study consists of a histomorphometric analysis of sections of iliac bone specimens from 35 women (age 16-78 years). First, the study shows that the age-induced cortical porosity reflects an increased pore size rather than an increased pore density. Second, it establishes a novel histomorphometric classification of the pores, which is based on the characteristics of the remodeling sites to which each pore is associated. It takes into consideration (i) the stage of the remodeling event at the level where the pore is sectioned, (ii) whether the event corresponds with the generation of a new pore through penetrative tunneling (type 1 pores) or with remodeling of an existing pore (type 2 pores), and (iii) in the latter case, whether or not the new remodeling event leads to the coalescence of pores. Of note, the advantage of this classification is to relate porosity with its generation mechanism. Third, it demonstrates that aging and porosity are correlated with: a shift from type 1 to type 2 pores, reflecting that the remodeling of existing pores is higher; an accumulation of eroded type 2 pores, reflecting an extended resorption-reversal phase; and a coalescence of these eroded type 2 pores into enlarged coalescing type 2 cavities. Collectively, this study supports the notion, that age-related increase in cortical porosity is the result of intracortical remodeling sites upon existing pores, with an extended reversal-resorption phase (eroded type 2 pores) that may likely result in a delayed or absent initiation of the subsequent bone formation. © 2017 The Authors. Journal of Bone and Mineral Research Published by Wiley Periodicals Inc.
Collapse
Affiliation(s)
- Christina Møller Andreasen
- Orthopeadic Research Laboratory, Department of Orthopaedic Surgery & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Jean-Marie Delaisse
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| | - Bram Cj van der Eerden
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Johannes Ptm van Leeuwen
- Laboratory for Calcium and Bone Metabolism, Department of Internal Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Ming Ding
- Orthopeadic Research Laboratory, Department of Orthopaedic Surgery & Traumatology, Odense University Hospital, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Institute of Regional Health Research, University of Southern Denmark, Vejle, Denmark
| |
Collapse
|
29
|
Ma Y, Yang H, Huang J. Icariin ameliorates dexamethasone‑induced bone deterioration in an experimental mouse model via activation of microRNA‑186 inhibition of cathepsin K. Mol Med Rep 2017; 17:1633-1641. [PMID: 29257214 PMCID: PMC5780104 DOI: 10.3892/mmr.2017.8065] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Accepted: 02/15/2017] [Indexed: 01/08/2023] Open
Abstract
The present study aimed to investigate bone deterioration in glucocorticoid‑induced osteoporosis (GIOP) mice, and the anti‑osteoporosis effect and underlying molecular mechanism of icariin. Dexamethasone (DSM) treatment was demonstrated to facilitate the induction of hypercalciuria in GIOP mice. Icariin treatment reversed the dexamethasone (DXM)‑induced disequilibrium of calcium homeostasis and bone resorption, and increased serum alkaline phosphatase, tartrate resistant acid phosphatase, osteocalcin and deoxypyridinoline. Haematoxylin and eosin staining revealed an increase in disconnections and separation in the trabecular bone network of the tibial proximal metaphysis, in the GIOP group. Icariin treatment reversed the DXM‑induced trabecular deleterious effects, and stimulated bone remodeling in GIOP mice. Furthermore, the results demonstrated that the mRNA and protein expression of cathepsin K were significantly increased in GIOP mice, compared with the control group. Icariin treatment may suppress the expression of cathepsin K in the tibia of GIOP mice. The levels of microRNA (miR)‑186 were markedly reduced in the tibia of GIOP mice compared with control group; however, this was inhibited by icariin treatment. Bioinformatics analysis demonstrated that miR‑186 regulates cathepsin K via binding to the upstream 3'‑untranslated region. Furthermore, transfection with miR‑186 mimics resulted in inhibition of cathepsin K expression, whereas miR‑186 inhibitors facilitated cathepsin K expression in osteoclasts. In conclusion, the present study demonstrated the protective effects of icariin against bone deteriorations in the experimental GIOP mice, and the underlying mechanism was mediated, at least partially, via activation of miR‑186‑mediated suppression of cathepsin K. These results provide evidence to support the use of icariin as a therapeutic approach in the management of glucocorticoid‑induced bone loss, and the disequilibrium of calcium homeostasis.
Collapse
Affiliation(s)
- Yongsheng Ma
- Department of Orthopaedics, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450002, P.R. China
| | - Hao Yang
- Department of Orthopaedics, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450002, P.R. China
| | - Junqing Huang
- Department of Orthopaedics, The Second Affiliated Hospital of Henan University of Traditional Chinese Medicine, Zhengzhou, Henan 450002, P.R. China
| |
Collapse
|
30
|
Saul D, Ninkovic M, Komrakova M, Wolff L, Simka P, Gasimov T, Menger B, Hoffmann DB, Rohde V, Sehmisch S. Effect of zileuton on osteoporotic bone and its healing, expression of bone, and brain genes in rats. J Appl Physiol (1985) 2017; 124:118-130. [PMID: 28860177 DOI: 10.1152/japplphysiol.01126.2016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Estrogen deficiency and aging are associated with osteoporosis, impaired bone healing, and lower cognitive performance. Close functional and physical connections occur between bone and the central nervous system. An anti-inflammatory drug, zileuton (which is an inhibitor of arachidonate 5-lipoxygenase), is known to have a positive effect on bone tissue repair and brain ischemia. We studied the effect of zileuton on osteopenic bone and its healing and on the genes considered to be crucial for the cross talks between bone and brain. Three-month-old Sprague-Dawley rats were ovariectomized or left untreated. After 8 wk, bilateral metaphyseal tibia osteotomy with plate osteosynthesis was performed in all rats. Ovariectomized rats were fed with food containing zileuton (1, 10, or 100 mg/kg body wt) for 5 wk. In tibiae, bone volume, callus and cortical volume, and gene expression of osteocalcin and alkaline phosphatase were enhanced by zileuton (10 or 100 mg); biomechanical properties and bone density were not changed. In femur, zileuton enlarged cortical volume distal and trabecular volume proximal, decreasing their density. The expression level of brain Sema3a, known to regulate bone mass positively, was downregulated after ovariectomy. In contrast, bone Sema4d, a negative regulator of bone mass, was upregulated in the tibia callus after ovariectomy, whereas zileuton treatment (10 or 100 mg) resulted in reverse effects. Here, we describe for the first time the expression of Rbbp4 mRNA and its increase in tibia after ovariectomy. Zileuton caused downregulation of Rbbp4 in the hippocampus and had an effect on bone healing, changed the expression of genes involved in cross talk between bones and brain, and may be a potent drug for further examination in estrogen deficiency-related dysfunction(s). NEW & NOTEWORTHY Zileuton, a 5-lipoxygenase inhibitor, increased bone volume, callus and cortical volume in osteotomized tibia, and trabecular and cortical volume in femur. Although the expression of Sema3a (positively regulating bone mass) in brain was downregulated and Sema4d (negatively regulating bone mass) was upregulated in tibia callus after ovariectomy, zileuton could counteract these effects. Rbbp4 (involved in age-related memory loss) was increased in tibia callus after ovariectomy.
Collapse
Affiliation(s)
- D Saul
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| | - M Ninkovic
- Department of Neurosurgery, University Medical Center Göttingen , Göttingen , Germany
| | - M Komrakova
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| | - L Wolff
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| | - P Simka
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| | - T Gasimov
- Department of Neurosurgery, University Medical Center Göttingen , Göttingen , Germany
| | - B Menger
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| | - D B Hoffmann
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| | - V Rohde
- Department of Neurosurgery, University Medical Center Göttingen , Göttingen , Germany
| | - S Sehmisch
- Department of Trauma Surgery, Orthopaedics and Plastic Surgery, University Medical Center Göttingen , Göttingen , Germany
| |
Collapse
|
31
|
Zhu S, Yao F, Qiu H, Zhang G, Xu H, Xu J. Coupling factors and exosomal packaging microRNAs involved in the regulation of bone remodelling. Biol Rev Camb Philos Soc 2017; 93:469-480. [PMID: 28795526 DOI: 10.1111/brv.12353] [Citation(s) in RCA: 90] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2016] [Revised: 06/18/2017] [Accepted: 06/26/2017] [Indexed: 12/31/2022]
Abstract
Bone remodelling is a continuous process by which bone resorption by osteoclasts is followed by bone formation by osteoblasts to maintain skeletal homeostasis. These two forces must be tightly coordinated not only quantitatively, but also in time and space, and its malfunction leads to diseases such as osteoporosis. Recent research focusing on the cross-talk and coupling mechanisms associated with the sequential recruitment of osteoblasts to areas where osteoclasts have removed bone matrix have identified a number of osteogenic factors produced by the osteoclasts themselves. Osteoclast-derived factors and exosomal-containing microRNA (miRNA) can either enhance or inhibit osteoblast differentiation through paracrine and juxtacrine mechanisms, and therefore may have a central coupling role in bone formation. Entwined with angiocrine factors released by vessel-specific endothelial cells and perivascular cells or pericytes, these factors play a critical role in angiogenesis-osteogenesis coupling essential in bone remodelling.
Collapse
Affiliation(s)
- Sipin Zhu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.,Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth 6009, M504, Australia
| | - Felix Yao
- Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth 6009, M504, Australia
| | - Heng Qiu
- Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth 6009, M504, Australia
| | - Ge Zhang
- Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Hong Kong SAR 999077, China
| | - Huazi Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Jiake Xu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, China.,Molecular Laboratory, School of Pathology and Laboratory Medicine, The University of Western Australia, Perth 6009, M504, Australia
| |
Collapse
|
32
|
Andreasen CM, Ding M, Andersen TL, Overgaard S. Effects of substitute coated with hyaluronic acid or poly‐lactic acid on implant fixation: Experimental study in ovariectomized and glucocorticoid‐treated sheep. J Tissue Eng Regen Med 2017; 12:e1122-e1130. [DOI: 10.1002/term.2447] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 02/16/2017] [Accepted: 05/04/2017] [Indexed: 12/12/2022]
Affiliation(s)
- Christina M. Andreasen
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology O, Odense University Hospital, Department of Clinical ResearchUniversity of Southern Denmark Odense Denmark
| | - Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology O, Odense University Hospital, Department of Clinical ResearchUniversity of Southern Denmark Odense Denmark
| | - Thomas L. Andersen
- Department of Clinical Cell Biology (KCB), Vejle Hospital – Lillebaelt Hospital, Department of Regional Health ResearchUniversity of Southern Denmark Vejle Denmark
| | - Søren Overgaard
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology O, Odense University Hospital, Department of Clinical ResearchUniversity of Southern Denmark Odense Denmark
| |
Collapse
|
33
|
Lassen NE, Andersen TL, Pløen GG, Søe K, Hauge EM, Harving S, Eschen GET, Delaisse JM. Coupling of Bone Resorption and Formation in Real Time: New Knowledge Gained From Human Haversian BMUs. J Bone Miner Res 2017; 32:1395-1405. [PMID: 28177141 DOI: 10.1002/jbmr.3091] [Citation(s) in RCA: 102] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 01/31/2023]
Abstract
It is well known that bone remodeling starts with a resorption event and ends with bone formation. However, what happens in between and how resorption and formation are coupled remains mostly unknown. Remodeling is achieved by so-called basic multicellular units (BMUs), which are local teams of osteoclasts, osteoblasts, and reversal cells recently proven identical with osteoprogenitors. Their organization within a BMU cannot be appropriately analyzed in common histology. The originality of the present study is to capture the events ranging from initiation of resorption to onset of formation as a functional continuum. It was based on the position of specific cell markers in longitudinal sections of Haversian BMUs generating new canals through human long bones. It showed that initial resorption at the tip of the canal is followed by a period where newly recruited reversal/osteoprogenitor cells and osteoclasts alternate, thus revealing the existence of a mixed "reversal-resorption" phase. Three-dimensional reconstructions obtained from serial sections indicated that initial resorption is mainly involved in elongating the canal and the additional resorption events in widening it. Canal diameter measurements show that the latter contribute the most to overall resorption. Of note, the density of osteoprogenitors continuously grew along the "reversal/resorption" surface, reaching at least 39 cells/mm on initiation of bone formation. This value was independent of the length of the reversal/resorption surface. These observations strongly suggest that bone formation is initiated only above a threshold cell density, that the length of the reversal/resorption period depends on how fast osteoprogenitor recruitment reaches this threshold, and thus that the slower the rate of osteoprogenitor recruitment, the more bone is degraded. They lead to a model where the newly recognized reversal/resorption phase plays a central role in the mechanism linking osteoprogenitor recruitment and the resorption-formation switch. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nicolai Ernlund Lassen
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | - Thomas Levin Andersen
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | - Gro Grunnet Pløen
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | - Kent Søe
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | | | - Søren Harving
- Department of Orthopaedic Surgery, Aalborg University Hospital, Aalborg, Denmark
| | | | - Jean-Marie Delaisse
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| |
Collapse
|
34
|
Paiva KBS, Granjeiro JM. Matrix Metalloproteinases in Bone Resorption, Remodeling, and Repair. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2017; 148:203-303. [PMID: 28662823 DOI: 10.1016/bs.pmbts.2017.05.001] [Citation(s) in RCA: 143] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Matrix metalloproteinases (MMPs) are the major protease family responsible for the cleavage of the matrisome (global composition of the extracellular matrix (ECM) proteome) and proteins unrelated to the ECM, generating bioactive molecules. These proteins drive ECM remodeling, in association with tissue-specific and cell-anchored inhibitors (TIMPs and RECK, respectively). In the bone, the ECM mediates cell adhesion, mechanotransduction, nucleation of mineralization, and the immobilization of growth factors to protect them from damage or degradation. Since the first description of an MMP in bone tissue, many other MMPs have been identified, as well as their inhibitors. Numerous functions have been assigned to these proteins, including osteoblast/osteocyte differentiation, bone formation, solubilization of the osteoid during bone resorption, osteoclast recruitment and migration, and as a coupling factor in bone remodeling under physiological conditions. In turn, a number of pathologies, associated with imbalanced bone remodeling, arise mainly from MMP overexpression and abnormalities of the ECM, leading to bone osteolysis or bone formation. In this review, we will discuss the functions of MMPs and their inhibitors in bone cells, during bone remodeling, pathological bone resorption (osteoporosis and bone metastasis), bone repair/regeneration, and emergent roles in bone bioengineering.
Collapse
Affiliation(s)
- Katiucia B S Paiva
- Laboratory of Extracellular Matrix Biology and Cellular Interaction (LabMec), Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil.
| | - José M Granjeiro
- National Institute of Metrology, Quality and Technology (InMetro), Bioengineering Laboratory, Duque de Caxias, RJ, Brazil; Fluminense Federal University, Dental School, Niterói, RJ, Brazil
| |
Collapse
|
35
|
Lv L, Ge W, Liu Y, Lai G, Liu H, Li W, Zhou Y. Lysine-specific demethylase 1 inhibitor rescues the osteogenic ability of mesenchymal stem cells under osteoporotic conditions by modulating H3K4 methylation. Bone Res 2016; 4:16037. [PMID: 28058134 PMCID: PMC5192052 DOI: 10.1038/boneres.2016.37] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 08/24/2016] [Accepted: 08/24/2016] [Indexed: 12/24/2022] Open
Abstract
Bone tissue engineering may be hindered by underlying osteoporosis because of a decreased osteogenic ability of autologous seed cells and an unfavorably changed microenvironment in these patients. Epigenetic regulation plays an important role in the developmental origins of osteoporosis; however, few studies have investigated the potential of epigenetic therapy to improve or rescue the osteogenic ability of bone marrow mesenchymal stem cells (BMMSCs) under osteoporotic conditions. Here, we investigated pargyline, an inhibitor of lysine-specific demethylase 1 (LSD1), which mainly catalyzes the demethylation of the di- and mono-methylation of H3K4. We demonstrated that 1.5 mmol·L−1 pargyline was the optimal concentration for the osteogenic differentiation of human BMMSCs. Pargyline rescued the osteogenic differentiation ability of mouse BMMSCs under osteoporotic conditions by enhancing the dimethylation level of H3K4 at the promoter regions of osteogenesis-related genes. Moreover, pargyline partially rescued or prevented the osteoporotic conditions in aged or ovariectomized mouse models, respectively. By introducing the concept of epigenetic therapy into the field of osteoporosis, this study demonstrated that LSD1 inhibitors could improve the clinical practice of MSC-based bone tissue engineering and proposes their novel use to treat osteoporosis.
Collapse
Affiliation(s)
- Longwei Lv
- Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing 100081, China
| | - Wenshu Ge
- Department of General Dentistry II, Peking University School and Hospital of Stomatology , Beijing 100081, China
| | - Yunsong Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing 100081, China
| | - Guanyou Lai
- Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing 100081, China
| | - Hao Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing 100081, China
| | - Wenyue Li
- Department of Prosthodontics, Peking University School and Hospital of Stomatology , Beijing 100081, China
| | - Yongsheng Zhou
- Department of Prosthodontics, Peking University School and Hospital of Stomatology, Beijing 100081, China; National Engineering Laboratory for Digital and Material Technology of Stomatology, Beijing 100081, China
| |
Collapse
|
36
|
Conaway HH, Henning P, Lie A, Tuckermann J, Lerner UH. Activation of dimeric glucocorticoid receptors in osteoclast progenitors potentiates RANKL induced mature osteoclast bone resorbing activity. Bone 2016; 93:43-54. [PMID: 27596806 DOI: 10.1016/j.bone.2016.08.024] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 08/26/2016] [Accepted: 08/30/2016] [Indexed: 01/02/2023]
Abstract
Glucocorticoid (GC) therapy is the greatest risk factor for secondary osteoporosis. Pathogenic mechanisms involve an initial increase in bone resorption followed by decreased bone formation. To gain a better understanding of the resorptive activity of GCs, we have used mouse bone marrow macrophages (BMM) to determine if GCs can directly modulate RANKL stimulated osteoclast formation and/or activity. In agreement with previous studies, experiments performed in plastic wells showed that GCs (dexamethasone, hydrocortisone, and prednisolone) inhibited osteoclast number and size during the initial phases of RANKL stimulated osteoclastogenesis; however, in prolonged cultures, decreased apoptosis was observed and escape from GC induced inhibition occurred with an enhanced number of osteoclasts formed, many with an increased area. When BMM cells were seeded on bone slices, GCs robustly enhanced RANKL stimulated formation of resorption pits and release of CTX without affecting the number or size of osteoclasts formed and with no effect on apoptosis. Stimulation of pit formation was not associated with increased life span of osteoclasts or an effect on mRNA expression of several osteoclastic or osteoclastogenic genes. The potentiation of RANKL induced CTX release by dexamethasone was significantly less in BMM cells from mice with conditional knockout of the osteoclastic glucocorticoid receptor and completely absent in cells from GRdim mice, which carry a point mutation in one dimerizing interface of the GC receptor. These data suggest that: 1. Plastic is a poor medium to use for studying direct effects of GCs on osteoclasts 2. GCs can enhance bone resorption without decreasing apoptosis, and 3. A direct enhancement of RANKL mediated resorption is stimulated by the dimeric GC-receptor.
Collapse
Affiliation(s)
- H Herschel Conaway
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, United States
| | - Petra Henning
- Centre for Bone and Arthritis Research at Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | - Anita Lie
- Department of Molecular Periodontology, Umeå University, Umeå, Sweden
| | - Jan Tuckermann
- Institute of Comparative Molecular Endocrinology, University of Ulm, Ulm, Germany
| | - Ulf H Lerner
- Centre for Bone and Arthritis Research at Department of Internal Medicine and Clinical Nutrition, Institute for Medicine, Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden; Department of Molecular Periodontology, Umeå University, Umeå, Sweden.
| |
Collapse
|
37
|
Pereira RC, Andersen TL, Friedman PA, Tumber N, Salusky IB, Wesseling-Perry K. Bone Canopies in Pediatric Renal Osteodystrophy. PLoS One 2016; 11:e0152871. [PMID: 27045269 PMCID: PMC4821625 DOI: 10.1371/journal.pone.0152871] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2015] [Accepted: 03/21/2016] [Indexed: 01/11/2023] Open
Abstract
Pediatric renal osteodystrophy (ROD) is characterized by changes in bone turnover, mineralization, and volume that are brought about by alterations in bone resorption and formation. The resorptive and formative surfaces on the cancellous bone are separated from the marrow cavity by canopies consisting of a layer of flat osteoblastic cells. These canopies have been suggested to play a key role in the recruitment of osteoprogenitors during the process of bone remodeling. This study was performed to address the characteristics of the canopies above bone formation and resorption sites and their association with biochemical and bone histomorphometric parameters in 106 pediatric chronic kidney disease (CKD) patients (stage 2-5) across the spectrum of ROD. Canopies in CKD patients often appeared as thickened multilayered canopies, similar to previous reports in patients with primary hyperparathyroidism. This finding contrasts with the thin appearance reported in healthy individuals with normal kidney function. Furthermore, canopies in pediatric CKD patients showed immunoreactivity to the PTH receptor (PTHR1) as well as to the receptor activator of nuclear factor kappa-B ligand (RANKL). The number of surfaces with visible canopy coverage was associated with plasma parathyroid hormone (PTH) levels, bone formation rate, and the extent of remodeling surfaces. Collectively, these data support the conclusion that canopies respond to the elevated PTH levels in CKD and that they possess the molecular machinery necessary to respond to PTH signaling.
Collapse
Affiliation(s)
- Renata C. Pereira
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Thomas L. Andersen
- Department of Clinical Cell Biology (KCB), Vejle Hospital – Lillebaelt Hospital, Institute of Regional Health Science, University of Southern Denmark, Vejle, Denmark
| | - Peter A. Friedman
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Navdeep Tumber
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Isidro B. Salusky
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Katherine Wesseling-Perry
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
38
|
Abdelgawad ME, Delaisse JM, Hinge M, Jensen PR, Alnaimi RW, Rolighed L, Engelholm LH, Marcussen N, Andersen TL. Early reversal cells in adult human bone remodeling: osteoblastic nature, catabolic functions and interactions with osteoclasts. Histochem Cell Biol 2016; 145:603-15. [PMID: 26860863 DOI: 10.1007/s00418-016-1414-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2015] [Indexed: 12/31/2022]
Abstract
The mechanism coupling bone resorption and formation is a burning question that remains incompletely answered through the current investigations on osteoclasts and osteoblasts. An attractive hypothesis is that the reversal cells are likely mediators of this coupling. Their nature is a big matter of debate. The present study performed on human cancellous bone is the first one combining in situ hybridization and immunohistochemistry to demonstrate their osteoblastic nature. It shows that the Runx2 and CD56 immunoreactive reversal cells appear to take up TRAcP released by neighboring osteoclasts. Earlier preclinical studies indicate that reversal cells degrade the organic matrix left behind by the osteoclasts and that this degradation is crucial for the initiation of the subsequent bone formation. To our knowledge, this study is the first addressing these catabolic activities in adult human bone through electron microscopy and analysis of molecular markers. Periosteoclastic reversal cells show direct contacts with the osteoclasts and with the demineralized resorption debris. These early reversal cells show (1) ¾-collagen fragments typically generated by extracellular collagenases of the MMP family, (2) MMP-13 (collagenase-3) and (3) the endocytic collagen receptor uPARAP/Endo180. The prevalence of these markers was lower in the later reversal cells, which are located near the osteoid surfaces and morphologically resemble mature bone-forming osteoblasts. In conclusion, this study demonstrates that reversal cells colonizing bone surfaces right after resorption are osteoblast-lineage cells, and extends to adult human bone remodeling their role in rendering eroded surfaces osteogenic.
Collapse
Affiliation(s)
- Mohamed Essameldin Abdelgawad
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.,Faculty of Science, Helwan University, Helwan, Egypt
| | - Jean-Marie Delaisse
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.
| | - Maja Hinge
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.,Division of Hematology, Department of Internal Medicine, Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Vejle, Denmark
| | - Pia Rosgaard Jensen
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark
| | - Ragad Walid Alnaimi
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark
| | - Lars Rolighed
- Breast and Endocrine Section, Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Lars H Engelholm
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Niels Marcussen
- Department of Clinical Pathology, Odense University Hospital, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.
| |
Collapse
|
39
|
Reagan MR, Rosen CJ. Navigating the bone marrow niche: translational insights and cancer-driven dysfunction. Nat Rev Rheumatol 2015; 12:154-68. [PMID: 26607387 DOI: 10.1038/nrrheum.2015.160] [Citation(s) in RCA: 92] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The bone marrow niche consists of stem and progenitor cells destined to become mature cells such as haematopoietic elements, osteoblasts or adipocytes. Marrow cells, influenced by endocrine, paracrine and autocrine factors, ultimately function as a unit to regulate bone remodelling and haematopoiesis. Current evidence highlights that the bone marrow niche is not merely an anatomic compartment; rather, it integrates the physiology of two distinct organ systems, the skeleton and the marrow. The niche has a hypoxic microenvironment that maintains quiescent haematopoietic stem cells (HSCs) and supports glycolytic metabolism. In response to biochemical cues and under the influence of neural, hormonal, and biochemical factors, marrow stromal elements, such as mesenchymal stromal cells (MSCs), differentiate into mature, functioning cells. However, disruption of the niche can affect cellular differentiation, resulting in disorders ranging from osteoporosis to malignancy. In this Review, we propose that the niche reflects the vitality of two tissues - bone and blood - by providing a unique environment for stem and stromal cells to flourish while simultaneously preventing disproportionate proliferation, malignant transformation or loss of the multipotent progenitors required for healing, functional immunity and growth throughout an organism's lifetime. Through a fuller understanding of the complexity of the niche in physiologic and pathologic states, the successful development of more-effective therapeutic approaches to target the niche and its cellular components for the treatment of rheumatic, endocrine, neoplastic and metabolic diseases becomes achievable.
Collapse
Affiliation(s)
- Michaela R Reagan
- Center for Molecular Medicine, Maine Medical Centre Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| | - Clifford J Rosen
- Center for Molecular Medicine, Maine Medical Centre Research Institute, 81 Research Drive, Scarborough, Maine 04074, USA
| |
Collapse
|
40
|
Reagan MR, Liaw L, Rosen CJ, Ghobrial IM. Dynamic interplay between bone and multiple myeloma: emerging roles of the osteoblast. Bone 2015; 75:161-9. [PMID: 25725265 PMCID: PMC4580250 DOI: 10.1016/j.bone.2015.02.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 02/15/2015] [Accepted: 02/18/2015] [Indexed: 01/06/2023]
Abstract
Multiple myeloma is a B-cell malignancy characterized by the unrelenting proliferation of plasma cells. Multiple myeloma causes osteolytic lesions and fractures that do not heal due to decreased osteoblastic and increased osteoclastic activity. However, the exact relationship between osteoblasts and myeloma cells remains elusive. Understanding the interactions between these dynamic bone-forming cells and myeloma cells is crucial to understanding how osteolytic lesions form and persist and how tumors grow within the bone marrow. This review provides a comprehensive overview of basic and translational research focused on the role of osteoblasts in multiple myeloma progression and their relationship to osteolytic lesions. Importantly, current challenges for in vitro studies exploring direct osteoblastic effects on myeloma cells, and gaps in understanding the role of the osteoblast in myeloma progression are delineated. Finally, successes and challenges in myeloma treatment with osteoanabolic therapy (i.e., any treatment that induces increased osteoblastic number or activity) are enumerated. Our goal is to illuminate novel mechanisms by which osteoblasts may contribute to multiple myeloma disease progression and osteolysis to better direct research efforts. Ultimately, we hope this may provide a roadmap for new approaches to the pathogenesis and treatment of multiple myeloma with a particular focus on the osteoblast.
Collapse
Affiliation(s)
- Michaela R Reagan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA; Maine Medical Center Research Institute, Scarborough, ME, USA
| | - Lucy Liaw
- Maine Medical Center Research Institute, Scarborough, ME, USA; Tufts University School of Medicine, Boston, MA, USA
| | - Clifford J Rosen
- Maine Medical Center Research Institute, Scarborough, ME, USA; Tufts University School of Medicine, Boston, MA, USA.
| | - Irene M Ghobrial
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
41
|
Sims NA, Martin TJ. Coupling Signals between the Osteoclast and Osteoblast: How are Messages Transmitted between These Temporary Visitors to the Bone Surface? Front Endocrinol (Lausanne) 2015; 6:41. [PMID: 25852649 PMCID: PMC4371744 DOI: 10.3389/fendo.2015.00041] [Citation(s) in RCA: 124] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 03/10/2015] [Indexed: 01/19/2023] Open
Affiliation(s)
- Natalie A. Sims
- Department of Medicine, St. Vincent’s Institute of Medical Research, St. Vincent’s Hospital, The University of Melbourne, Fitzroy, VIC, Australia
- *Correspondence:
| | - T. John Martin
- Department of Medicine, St. Vincent’s Institute of Medical Research, St. Vincent’s Hospital, The University of Melbourne, Fitzroy, VIC, Australia
| |
Collapse
|