1
|
Moura SR, Sousa AB, Olesen JB, Barbosa MA, Søe K, Almeida MI. Stage-specific modulation of multinucleation, fusion, and resorption by the long non-coding RNA DLEU1 and miR-16 in human primary osteoclasts. Cell Death Dis 2024; 15:741. [PMID: 39389940 PMCID: PMC11467329 DOI: 10.1038/s41419-024-06983-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 08/01/2024] [Accepted: 08/07/2024] [Indexed: 10/12/2024]
Abstract
Osteoclasts are the only cells able to resorb all the constituents of the bone matrix. While the modulation of osteoclast activity is well established for preventing bone-related diseases, there is an increasing demand for novel classes of anti-resorption agents. Herein, we investigated non-coding RNA molecules and proposed DLEU1 and miR-16 as potential candidates for modulating osteoclast functions. DLEU1 and miR-16 target cell fusion at both the early and late stages of osteoclastogenesis but operate through independent pathways. DLEU1 silencing hinders the fusion process, leading to abrogation of the phagocytic cup fusion modality and a reduction in the fusion events between mononucleated precursors and multinucleated osteoclasts, while miR-16 influences monocyte-to-osteoclast differentiation, impairing osteoclasts formation but not the number of nuclei at early stages. On the other hand, using these non-coding RNAs to engineer mature osteoclasts has implications for bone resorption. Both DLEU1 and miR-16 influence the speed of resorption in pit-forming osteoclasts, without affecting the resorbed area. However, the impact of increasing miR-16 levels extends more broadly, affecting trench-forming osteoclasts as well, leading to a reduction in their percentage, speed, and resorbed area. These findings offer potential new therapeutic targets to ameliorate bone destruction in skeletal diseases.
Collapse
Affiliation(s)
- Sara Reis Moura
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Beatriz Sousa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Jacob Bastholm Olesen
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Mário Adolfo Barbosa
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Kent Søe
- Department of Pathology, Odense University Hospital, Odense, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Maria Inês Almeida
- i3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
- INEB-Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
2
|
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
|
3
|
Pandit A, Indurkar A, Locs J, Haugen HJ, Loca D. Calcium Phosphates: A Key to Next-Generation In Vitro Bone Modeling. Adv Healthc Mater 2024:e2401307. [PMID: 39175382 DOI: 10.1002/adhm.202401307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 08/06/2024] [Indexed: 08/24/2024]
Abstract
The replication of bone physiology under laboratory conditions is a prime target behind the development of in vitro bone models. The model should be robust enough to elicit an unbiased response when stimulated experimentally, giving reproducible outcomes. In vitro bone tissue generation majorly requires the availability of cellular components, the presence of factors promoting cellular proliferation and differentiation, efficient nutrient supply, and a supporting matrix for the cells to anchor - gaining predefined topology. Calcium phosphates (CaP) are difficult to ignore while considering the above requirements of a bone model. Therefore, the current review focuses on the role of CaP in developing an in vitro bone model addressing the prerequisites of bone tissue generation. Special emphasis is given to the physico-chemical properties of CaP that promote osteogenesis, angiogenesis and provide sufficient mechanical strength for load-bearing applications. Finally, the future course of action is discussed to ensure efficient utilization of CaP in the in vitro bone model development field.
Collapse
Affiliation(s)
- Ashish Pandit
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| | - Abhishek Indurkar
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| | - Janis Locs
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| | | | - Dagnija Loca
- Institute of Biomaterials and Bioengineering, Faculty of Natural Sciences and Technology, Riga Technical University, Pulka Street 3, Riga, LV-1007, Latvia
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Riga, LV-1007, Latvia
| |
Collapse
|
4
|
Yoon H, Park Y, Kwak JG, Lee J. Collagen structures of demineralized bone paper direct mineral metabolism. JBMR Plus 2024; 8:ziae080. [PMID: 38989259 PMCID: PMC11235081 DOI: 10.1093/jbmrpl/ziae080] [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: 12/18/2023] [Revised: 05/28/2024] [Accepted: 06/12/2024] [Indexed: 07/12/2024] Open
Abstract
Bone is a dynamic mineralized tissue that undergoes continuous turnover throughout life. While the general mechanism of bone mineral metabolism is documented, the role of underlying collagen structures in regulating osteoblastic mineral deposition and osteoclastic mineral resorption remains an active research area, partly due to the lack of biomaterial platforms supporting accurate and analytical investigation. The recently introduced osteoid-inspired demineralized bone paper (DBP), prepared by 20-μm thin sectioning of demineralized bovine compact bone, holds promise in addressing this challenge as it preserves the intrinsic bony collagen structure and retains semi-transparency. Here, we report on the impact of collagen structures on modulating osteoblast and osteoclast-driven bone mineral metabolism using vertical and transversal DBPs that exhibit a uniaxially aligned and a concentric ring collagen structure, respectively. Translucent DBP reveals these collagen structures and facilitates longitudinal tracking of mineral deposition and resorption under brightfield microscopy for at least 3 wk. Genetically labeled primary osteogenic cells allow fluorescent monitoring of these cellular processes. Osteoblasts adhere and proliferate following the underlying collagen structures of DBPs. Osteoblastic mineral deposition is significantly higher in vertical DBP than in transversal DBP. Spatiotemporal analysis reveals notably more osteoblast adhesion and faster mineral deposition in vascular regions than in bone regions. Subsequent osteoclastic resorption follows these mineralized collagen structures, directing distinct trench and pit-type resorption patterns. In vertical DBP, trench-type resorption occurs at an 80% frequency, whereas transversal DBP shows 35% trench-type and 65% pit-type resorption. Our studies substantiate the importance of collagen structures in regulating mineral metabolism by osteogenic cells. DBP is expected to serve as an enabling biomaterial platform for studying various aspects of cellular and extracellular bone remodeling biology.
Collapse
Affiliation(s)
- Hyejin Yoon
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, MA 01003, United States
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, United States
| | - Jun-Goo Kwak
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
| | - Jungwoo Lee
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, United States
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, United States
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, United States
| |
Collapse
|
5
|
Grass DM, Malek G, Taïeb HM, Ittah E, Richard H, Reznikov N, Laverty S. Characterization and quantification of in-vitro equine bone resorption in 3D using μCT and deep learning-aided feature segmentation. Bone 2024; 185:117131. [PMID: 38777311 DOI: 10.1016/j.bone.2024.117131] [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: 02/08/2024] [Revised: 05/18/2024] [Accepted: 05/19/2024] [Indexed: 05/25/2024]
Abstract
High cyclic strains induce formation of microcracks in bone, triggering targeted bone remodeling, which entails osteoclastic resorption. Racehorse bone is an ideal model for studying the effects of high-intensity loading, as it is subject to focal formation of microcracks and subsequent bone resorption. The volume of resorption in vitro is considered a direct indicator of osteoclast activity but indirect 2D measurements are used more often. Our objective was to develop an accurate, high-throughput method to quantify equine osteoclast resorption volume in μCT 3D images. Here, equine osteoclasts were cultured on equine bone slices and imaged with μCT pre- and postculture. Individual resorption events were then isolated and analyzed in 3D. Modal volume, maximum depth, and aspect ratio of resorption events were calculated. A convolutional neural network (CNN U-Net-like) was subsequently trained to identify resorption events on post-culture μCT images alone, without the need for pre-culture imaging, using archival bone slices with known resorption areas and paired CTX-I biomarker levels in culture media. 3D resorption volume measurements strongly correlated with both the CTX-I levels (p < 0.001) and area measurements (p < 0.001). Our 3D analysis shows that the shapes of resorption events form a continuous spectrum, rather than previously reported pit and trench categories. With more extensive resorption, shapes of increasing complexity appear, although simpler resorption cavity morphologies (small, rounded) remain most common, in acord with the left-hand limit paradigm. Finally, we show that 2D measurements of in vitro osteoclastic resorption are a robust and reliable proxy.
Collapse
Affiliation(s)
- Debora M Grass
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada
| | - Gwladys Malek
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada
| | - Hubert M Taïeb
- Department of Bioengineering, Faculty of Engineering, McGill University, 3480 University Street, Montreal, Quebec H3A 0E9, Canada
| | - Eran Ittah
- Department of Bioengineering, Faculty of Engineering, McGill University, 3480 University Street, Montreal, Quebec H3A 0E9, Canada
| | - Hélène Richard
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada
| | - Natalie Reznikov
- Department of Bioengineering, Faculty of Engineering, McGill University, 3480 University Street, Montreal, Quebec H3A 0E9, Canada
| | - Sheila Laverty
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC J2S 2M2, Canada.
| |
Collapse
|
6
|
Nielsen SSR, Pedersen JAZ, Sharma N, Wasehuus PK, Hansen MS, Møller AMJ, Borggaard XG, Rauch A, Frost M, Sondergaard TE, Søe K. Human osteoclasts in vitro are dose dependently both inhibited and stimulated by cannabidiol (CBD) and Δ9-tetrahydrocannabinol (THC). Bone 2024; 181:117035. [PMID: 38342278 DOI: 10.1016/j.bone.2024.117035] [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: 12/09/2023] [Revised: 01/12/2024] [Accepted: 01/31/2024] [Indexed: 02/13/2024]
Abstract
Legalized use of cannabis for medical or recreational use is becoming more and more common. With respect to potential side-effects on bone health only few clinical trials have been conducted - and with opposing results. Therefore, it seems that there is a need for more knowledge on the potential effects of cannabinoids on human bone cells. We studied the effect of cannabidiol (CBD) and Δ9-tetrahydrocannabinol (THC) (dose range from 0.3 to 30 μM) on human osteoclasts in mono- as well as in co-cultures with human osteoblast lineage cells. We have used CD14+ monocytes from anonymous blood donors to differentiate into osteoclasts, and human osteoblast lineage cells from outgrowths of human trabecular bone. Our results show that THC and CBD have dose-dependent effects on both human osteoclast fusion and bone resorption. In the lower dose ranges of THC and CBD, osteoclast fusion was unaffected while bone resorption was increased. At higher doses, both osteoclast fusion and bone resorption were inhibited. In co-cultures, both osteoclastic bone resorption and alkaline phosphatase activity of the osteoblast lineage cells were inhibited. Finally, we observed that the cannabinoid receptor CNR2 is more highly expressed than CNR1 in CD14+ monocytes and pre-osteoclasts, but also that differentiation to osteoclasts was coupled to a reduced expression of CNR2, in particular. Interestingly, under co-culture conditions, we only detected the expression of CNR2 but not CNR1 for both osteoclast as well as osteoblast lineage nuclei. In line with the existing literature on the effect of cannabinoids on bone cells, our current study shows both stimulatory and inhibitory effects. This highlights that potential unfavorable effects of cannabinoids on bone cells and bone health is a complex matter. The contradictory and lacking documentation for such potential unfavorable effects on bone health as well as other potential effects, should be taken into consideration when considering the use of cannabinoids for both medical and recreational use.
Collapse
Affiliation(s)
- Simone S R Nielsen
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Department of Pathology, Odense University Hospital, J.B. Winsløws Vej 15, 5000 Odense C, Denmark.
| | - Juliana A Z Pedersen
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Department of Pathology, Odense University Hospital, J.B. Winsløws Vej 15, 5000 Odense C, Denmark.
| | - Neha Sharma
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Department of Pathology, Odense University Hospital, J.B. Winsløws Vej 15, 5000 Odense C, Denmark; Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Pernille K Wasehuus
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220 Aalborg, Denmark
| | - Morten S Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, J.B. Winsløws Vej 4, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Anaïs M J Møller
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, Kabbeltoft 25, 7100 Vejle, Denmark.
| | - Xenia G Borggaard
- Department of Pathology, Odense University Hospital, J.B. Winsløws Vej 15, 5000 Odense C, Denmark; Molecular Bone Histology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| | - Alexander Rauch
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, J.B. Winsløws Vej 4, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Steno Diabetes Centre Odense, Odense University Hospital, Kløvervænget 10, 5000 Odense C, Denmark.
| | - Morten Frost
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, J.B. Winsløws Vej 4, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Steno Diabetes Centre Odense, Odense University Hospital, Kløvervænget 10, 5000 Odense C, Denmark.
| | - Teis E Sondergaard
- Department of Chemistry and Bioscience, Aalborg University, Fredrik Bajers Vej 7H, 9220 Aalborg, Denmark.
| | - Kent Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark; Department of Pathology, Odense University Hospital, J.B. Winsløws Vej 15, 5000 Odense C, Denmark; Department of Molecular Medicine, University of Southern Denmark, Campusvej 55, 5230 Odense M, Denmark.
| |
Collapse
|
7
|
Panwar P, Olesen JB, Blum G, Delaisse JM, Søe K, Brömme D. Real-time analysis of osteoclast resorption and fusion dynamics in response to bone resorption inhibitors. Sci Rep 2024; 14:7358. [PMID: 38548807 PMCID: PMC10978898 DOI: 10.1038/s41598-024-57526-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 03/19/2024] [Indexed: 04/01/2024] Open
Abstract
Cathepsin K (CatK), an essential collagenase in osteoclasts (OCs), is a potential therapeutic target for the treatment of osteoporosis. Using live-cell imaging, we monitored the bone resorptive behaviour of OCs during dose-dependent inhibition of CatK by an ectosteric (Tanshinone IIA sulfonate) and an active site inhibitor (odanacatib). CatK inhibition caused drastic reductions in the overall resorption speed of OCs. At IC50 CatK-inhibitor concentration, OCs reduced about 40% of their trench-forming capacity and at fourfold IC50 concentrations, a > 95% reduction was observed. The majority of CatK-inhibited OCs (~ 75%) were involved in resorption-migration-resorption episodes forming adjacent pits, while ~ 25% were stagnating OCs which remained associated with the same excavation. We also observed fusions of OCs during the resorption process both in control and inhibitor-treated conditions, which increased their resorption speeds by 30-50%. Inhibitor IC50-concentrations increased OC-fusion by twofold. Nevertheless, more fusion could not counterweigh the overall loss of resorption activity by inhibitors. Using an activity-based probe, we demonstrated the presence of active CatK at the resorbing front in pits and trenches. In conclusion, our data document how OCs respond to CatK-inhibition with respect to movement, bone resorption activity, and their attempt to compensate for inhibition by activating fusion.
Collapse
Affiliation(s)
- Preety Panwar
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada
- Department of Pharmaceutical Sciences, Elizabeth City State University, Elizabeth City, NC, USA
| | - Jacob Bastholm Olesen
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
- Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Galia Blum
- Faculty of Medicine, Campus Ein Karem, The School of Pharmacy, Institute of Drug Research, The Hebrew University of Jerusalem, Room 407, 9112001, Jerusalem, Israel
| | - Jean-Marie Delaisse
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark
| | - Kent Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Campusvej 55, 5230, Odense M, Denmark.
- Department of Pathology, Odense University Hospital, Odense, Denmark.
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| | - Dieter Brömme
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada.
- Department of Oral Biological and Medical Sciences, Faculty of Dentistry, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, V6T 1Z3, Canada.
| |
Collapse
|
8
|
Chandrabalan S, Dang L, Hansen U, Timmen M, Wehmeyer C, Stange R, Beißbarth T, Binder C, Bleckmann A, Menck K. A novel method to efficiently differentiate human osteoclasts from blood-derived monocytes. Biol Proced Online 2024; 26:7. [PMID: 38504200 PMCID: PMC10949786 DOI: 10.1186/s12575-024-00233-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Accepted: 03/09/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Osteoclasts are the tissue-specific macrophage population of the bone and unique in their bone-resorbing activity. Hence, they are fundamental for bone physiology in health and disease. However, efficient protocols for the isolation and study of primary human osteoclasts are scarce. In this study, we aimed to establish a protocol, which enables the efficient differentiation of functional human osteoclasts from monocytes. RESULTS Human monocytes were isolated through a double-density gradient from donor blood. Compared to standard differentiation schemes in polystyrene cell culture dishes, the yield of multinuclear osteoclasts was significantly increased upon initial differentiation of monocytes to macrophages in fluorinated ethylene propylene (FEP) Teflon bags. This initial differentiation phase was then followed by the development of terminal osteoclasts by addition of Receptor Activator of NF-κB Ligand (RANKL). High concentrations of RANKL and Macrophage colony-stimulating factor (M-CSF) as well as an intermediate cell density further supported efficient cell differentiation. The generated cells were highly positive for CD45, CD14 as well as the osteoclast markers CD51/ITGAV and Cathepsin K/CTSK, thus identifying them as osteoclasts. The bone resorption of the osteoclasts was significantly increased when the cells were differentiated from macrophages derived from Teflon bags compared to macrophages derived from conventional cell culture plates. CONCLUSION Our study has established a novel protocol for the isolation of primary human osteoclasts that improves osteoclastogenesis in comparison to the conventionally used cultivation approach.
Collapse
Affiliation(s)
- Suganja Chandrabalan
- Department of Medicine A, Hematology, Oncology, and Pneumology, University of Muenster, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, Muenster, Germany
| | - Linh Dang
- Department of Medical Bioinformatics, University Medical Center Goettingen, Goettingen, Germany
| | - Uwe Hansen
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Melanie Timmen
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Corinna Wehmeyer
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Richard Stange
- Institute of Musculoskeletal Medicine (IMM), University of Muenster, Muenster, Germany
| | - Tim Beißbarth
- Department of Medical Bioinformatics, University Medical Center Goettingen, Goettingen, Germany
| | - Claudia Binder
- Department of Hematology/Medical Oncology, University Medical Center Goettingen, Goettingen, Germany
| | - Annalen Bleckmann
- Department of Medicine A, Hematology, Oncology, and Pneumology, University of Muenster, Muenster, Germany
- West German Cancer Center, University Hospital Muenster, Muenster, Germany
| | - Kerstin Menck
- Department of Medicine A, Hematology, Oncology, and Pneumology, University of Muenster, Muenster, Germany.
- West German Cancer Center, University Hospital Muenster, Muenster, Germany.
| |
Collapse
|
9
|
Hansen MS, Madsen K, Price M, Søe K, Omata Y, Zaiss MM, Gorvin CM, Frost M, Rauch A. Transcriptional reprogramming during human osteoclast differentiation identifies regulators of osteoclast activity. Bone Res 2024; 12:5. [PMID: 38263167 PMCID: PMC10806178 DOI: 10.1038/s41413-023-00312-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 11/08/2023] [Accepted: 12/15/2023] [Indexed: 01/25/2024] Open
Abstract
Enhanced osteoclastogenesis and osteoclast activity contribute to the development of osteoporosis, which is characterized by increased bone resorption and inadequate bone formation. As novel antiosteoporotic therapeutics are needed, understanding the genetic regulation of human osteoclastogenesis could help identify potential treatment targets. This study aimed to provide an overview of transcriptional reprogramming during human osteoclast differentiation. Osteoclasts were differentiated from CD14+ monocytes from eight female donors. RNA sequencing during differentiation revealed 8 980 differentially expressed genes grouped into eight temporal patterns conserved across donors. These patterns revealed distinct molecular functions associated with postmenopausal osteoporosis susceptibility genes based on RNA from iliac crest biopsies and bone mineral density SNPs. Network analyses revealed mutual dependencies between temporal expression patterns and provided insight into subtype-specific transcriptional networks. The donor-specific expression patterns revealed genes at the monocyte stage, such as filamin B (FLNB) and oxidized low-density lipoprotein receptor 1 (OLR1, encoding LOX-1), that are predictive of the resorptive activity of mature osteoclasts. The expression of differentially expressed G-protein coupled receptors was strong during osteoclast differentiation, and these receptors are associated with bone mineral density SNPs, suggesting that they play a pivotal role in osteoclast differentiation and activity. The regulatory effects of three differentially expressed G-protein coupled receptors were exemplified by in vitro pharmacological modulation of complement 5 A receptor 1 (C5AR1), somatostatin receptor 2 (SSTR2), and free fatty acid receptor 4 (FFAR4/GPR120). Activating C5AR1 enhanced osteoclast formation, while activating SSTR2 decreased the resorptive activity of mature osteoclasts, and activating FFAR4 decreased both the number and resorptive activity of mature osteoclasts. In conclusion, we report the occurrence of transcriptional reprogramming during human osteoclast differentiation and identified SSTR2 and FFAR4 as antiresorptive G-protein coupled receptors and FLNB and LOX-1 as potential molecular markers of osteoclast activity. These data can help future investigations identify molecular regulators of osteoclast differentiation and activity and provide the basis for novel antiosteoporotic targets.
Collapse
Affiliation(s)
- Morten S Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000, Odense C, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, DK-5000, Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, DK-5000, Odense C, Denmark
| | - Kaja Madsen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000, Odense C, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, DK-5000, Odense C, Denmark
| | - Maria Price
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham, B15 2TT, UK
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, B15 2TT, UK
| | - Kent Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, DK-5000, Odense C, Denmark
- Department of Molecular Medicine, University of Southern Denmark, DK-5000, Odense C, Denmark
| | - Yasunori Omata
- Department of Orthopedic Surgery, Faculty of Medicine, The University of Tokyo, Tokyo, 113-8655, Japan
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, D-91054, Erlangen, Germany
| | - Mario M Zaiss
- Department of Internal Medicine 3, Rheumatology and Immunology, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) and Universitätsklinikum Erlangen, D-91054, Erlangen, Germany
- Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, D-91054, Erlangen, Germany
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham, B15 2TT, UK
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, B15 2TT, UK
| | - Morten Frost
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000, Odense C, Denmark.
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, DK-5000, Odense C, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000, Odense C, Denmark.
| | - Alexander Rauch
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, DK-5000, Odense C, Denmark.
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, DK-5000, Odense C, Denmark.
- Steno Diabetes Center Odense, Odense University Hospital, DK-5000, Odense C, Denmark.
| |
Collapse
|
10
|
Ozanne H, Moubri L, Abou-Nassif L, Thoumire O, Echalard A, Morin-Grognet S, Atmani H, Ladam G, Labat B. Active Osteoblasts or Quiescent Bone Lining Cells? Preosteoblasts Fate Orchestrated by Curvature and Stiffness of an In Vitro 2.5D Biomimetic Culture System. Adv Healthc Mater 2024; 13:e2302222. [PMID: 37929897 DOI: 10.1002/adhm.202302222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Indexed: 11/07/2023]
Abstract
Biomimetic cell culture systems are required to provide more physiologically relevant microenvironments for bone cells. Here, a simple 2.5D culture platform is proposed, combining adjustable stiffness and surface features that mimic bone topography by using sandpaper grits as master molds with two stiffness formulations of polydimethylsiloxane (PDMS). The subsequent replicas perfectly conform the grits and reproduce the corresponding negative relief with cavities separated by convex edges. Biomimicry is also provided by an extracellular matrix (ECM)-like thin film coating, using the layer-by-layer (LbL) method. The topographical features, alternating concave, and convex structures drive preosteoblasts organization and morphology. Strikingly, curvature orchestrates the commitment of preosteoblasts, with i) maturation to active osteoblasts able to produce a dense collagenous matrix that ultimately mineralizes in the cavities, and ii) edges hosting quiescent cells that synthetize a very thin immature collagen layer with no mineralization. In summary, the present in vitro culture system model offers a cell-instructive 2.5D microenvironment that controls preosteoblasts fate, leading to two coexisting subpopulations: mature osteoblasts and bone lining cells (BLC). This promising culture system opens new avenues to advanced tissue-engineered modeling and can be applied to precellularized bone biomaterials.
Collapse
Affiliation(s)
- Hélène Ozanne
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Loïc Moubri
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Léa Abou-Nassif
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Olivier Thoumire
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Aline Echalard
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | | | - Hassan Atmani
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Guy Ladam
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| | - Béatrice Labat
- Univ Rouen Normandie, INSA Rouen Normandie, CNRS, PBS UMR 6270, Evreux, F-27000, France
| |
Collapse
|
11
|
Cramer EEA, de Wildt BWM, Hendriks JGE, Ito K, Hofmann S. Integration of osteoclastogenesis through addition of PBMCs in human osteochondral explants cultured ex vivo. Bone 2024; 178:116935. [PMID: 37852425 DOI: 10.1016/j.bone.2023.116935] [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: 08/11/2023] [Revised: 09/29/2023] [Accepted: 10/10/2023] [Indexed: 10/20/2023]
Abstract
The preservation of tissue specific cells in their native 3D extracellular matrix in bone explants provides a unique platform to study remodeling. Thus far, studies involving bone explant cultures showed a clear focus on achieving bone formation and neglected osteoclast activity and resorption. To simulate the homeostatic bone environment ex vivo, both key elements of bone remodeling need to be represented. This study aimed to assess and include osteoclastogenesis in human osteochondral explants through medium supplementation with RANKL and M-CSF and addition of peripheral blood mononuclear cells (PBMCs), providing osteoclast precursors. Osteochondral explants were freshly harvested from human femoral heads obtained from hip surgeries and cultured for 20 days in a two-compartment culture system. Osteochondral explants preserved viability and cellular abundance over the culture period, but histology demonstrated that resident osteoclasts were no longer present after 4 days of culture. Quantitative extracellular tartrate resistant acid phosphatase (TRAP) analysis confirmed depletion of osteoclast activity on day 4 even when stimulated with RANKL and M-CSF. Upon addition of PBMCs, a significant upregulation of TRAP activity was measured from day 10 onwards. Evaluation of bone loss trough μCT registration and measurement of extracellular cathepsin K activity revealed indications of enhanced resorption upon addition of PBMCs. Based on the results we suggest that an external source of osteoclast precursors, such as PBMCs, needs to be added in long-term bone explant cultures to maintain osteoclastic activity, and bone remodeling.
Collapse
Affiliation(s)
- Esther E A Cramer
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Bregje W M de Wildt
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Johannes G E Hendriks
- Department of Orthopedic Surgery & Trauma, Máxima Medical Center Eindhoven/Veldhoven, 5631 BM Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute of Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB Eindhoven, the Netherlands.
| |
Collapse
|
12
|
Park Y, Sato T, Lee J. Functional and analytical recapitulation of osteoclast biology on demineralized bone paper. Nat Commun 2023; 14:8092. [PMID: 38062034 PMCID: PMC10703810 DOI: 10.1038/s41467-023-44000-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/27/2023] [Indexed: 12/18/2023] Open
Abstract
Osteoclasts are the primary target for osteoporosis drug development. Recent animal studies revealed the crucial roles of osteoblasts in regulating osteoclastogenesis and the longer lifespans of osteoclasts than previously thought with fission and recycling. However, existing culture platforms are limited to replicating these newly identified cellular processes. We report a demineralized bone paper (DBP)-based osteoblast culture and osteoclast assay platform that replicates osteoclast fusion, fission, resorption, and apoptosis with high fidelity and analytical power. An osteoid-inspired DBP supports rapid and structural mineral deposition by osteoblasts. Coculture osteoblasts and bone marrow monocytes under biochemical stimulation recapitulate osteoclast differentiation and function. The DBP-based bone model allows longitudinal quantitative fluorescent monitoring of osteoclast responses to bisphosphonate drug, substantiating significantly reducing their number and lifespan. Finally, we demonstrate the feasibility of humanizing the bone model. The DBP-based osteo assay platforms are expected to advance bone remodeling-targeting drug development with improved prediction of clinical outcomes.
Collapse
Affiliation(s)
- Yongkuk Park
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA
| | - Tadatoshi Sato
- Department of Medicine, UMass Chan Medical School, Worcester, MA, 01605, USA
| | - Jungwoo Lee
- Department of Chemical Engineering, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA, 01003, USA.
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01003, USA.
- Molecular & Cellular Biology Graduate Program, University of Massachusetts, Amherst, MA, 01003, USA.
| |
Collapse
|
13
|
Torres HM, Arnold KM, Oviedo M, Westendorf JJ, Weaver SR. Inflammatory Processes Affecting Bone Health and Repair. Curr Osteoporos Rep 2023; 21:842-853. [PMID: 37759135 PMCID: PMC10842967 DOI: 10.1007/s11914-023-00824-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2023] [Indexed: 09/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this article is to review the current understanding of inflammatory processes on bone, including direct impacts of inflammatory factors on bone cells, the effect of senescence on inflamed bone, and the critical role of inflammation in bone pain and healing. RECENT FINDINGS Advances in osteoimmunology have provided new perspectives on inflammatory bone loss in recent years. Characterization of so-called inflammatory osteoclasts has revealed insights into physiological and pathological bone loss. The identification of inflammation-associated senescent markers in bone cells indicates that therapies that reduce senescent cell burden may reverse bone loss caused by inflammatory processes. Finally, novel studies have refined the role of inflammation in bone healing, including cross talk between nerves and bone cells. Except for the initial stages of fracture healing, inflammation has predominately negative effects on bone and increases fracture risk. Eliminating senescent cells, priming the osteo-immune axis in bone cells, and alleviating pro-inflammatory cytokine burden may ameliorate the negative effects of inflammation on bone.
Collapse
Affiliation(s)
- Haydee M Torres
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Katherine M Arnold
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Biomedical Engineering and Physiology Track/Regenerative Sciences Program, Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN, 55905, USA
| | - Manuela Oviedo
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Jennifer J Westendorf
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Samantha R Weaver
- Department of Orthopedic Surgery, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
14
|
Davies BK, Hibbert AP, Roberts SJ, Roberts HC, Tickner JC, Holdsworth G, Arnett TR, Orriss IR. A Machine Learning-Based Image Segmentation Method to Quantify In Vitro Osteoclast Culture Endpoints. Calcif Tissue Int 2023; 113:437-448. [PMID: 37566229 PMCID: PMC10516805 DOI: 10.1007/s00223-023-01121-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 07/29/2023] [Indexed: 08/12/2023]
Abstract
Quantification of in vitro osteoclast cultures (e.g. cell number) often relies on manual counting methods. These approaches are labour intensive, time consuming and result in substantial inter- and intra-user variability. This study aimed to develop and validate an automated workflow to robustly quantify in vitro osteoclast cultures. Using ilastik, a machine learning-based image analysis software, images of tartrate resistant acid phosphatase-stained mouse osteoclasts cultured on dentine discs were used to train the ilastik-based algorithm. Assessment of algorithm training showed that osteoclast numbers strongly correlated between manual- and automatically quantified values (r = 0.87). Osteoclasts were consistently faithfully segmented by the model when visually compared to the original reflective light images. The ability of this method to detect changes in osteoclast number in response to different treatments was validated using zoledronate, ticagrelor, and co-culture with MCF7 breast cancer cells. Manual and automated counting methods detected a 70% reduction (p < 0.05) in osteoclast number, when cultured with 10 nM zoledronate and a dose-dependent decrease with 1-10 μM ticagrelor (p < 0.05). Co-culture with MCF7 cells increased osteoclast number by ≥ 50% irrespective of quantification method. Overall, an automated image segmentation and analysis workflow, which consistently and sensitively identified in vitro osteoclasts, was developed. Advantages of this workflow are (1) significantly reduction in user variability of endpoint measurements (93%) and analysis time (80%); (2) detection of osteoclasts cultured on different substrates from different species; and (3) easy to use and freely available to use along with tutorial resources.
Collapse
Affiliation(s)
- Bethan K Davies
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
- Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Andrew P Hibbert
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Scott J Roberts
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
| | - Helen C Roberts
- Department of Natural Sciences, Middlesex University, London, UK
| | - Jennifer C Tickner
- School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | | | - Timothy R Arnett
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK
- Department of Cell and Developmental Biology, University College London, London, UK
| | - Isabel R Orriss
- Department of Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
15
|
Ghanta P, Winschel T, Hessel E, Oyewumi O, Czech T, Oyewumi MO. Efficacy assessment of methylcellulose-based thermoresponsive hydrogels loaded with gallium acetylacetonate in osteoclastic bone resorption. Drug Deliv Transl Res 2023; 13:2533-2549. [PMID: 37014587 PMCID: PMC10469133 DOI: 10.1007/s13346-023-01336-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2023] [Indexed: 04/05/2023]
Abstract
Homeostatic imbalance involving progressive stimulation of osteoclast (OC) differentiation and function will lead to an increased risk of fragility fractures. In this regard, we investigated gallium acetylacetonate (GaAcAc) as a possible treatment for osteoclastic bone resorption. Further, the extent to which suitable delivery systems can enhance the therapeutic potential of GaAcAc was evaluated. GaAcAc solution (10-50 µg/mL) suppressed OC differentiation using murine monocytic RAW 264.7 or hematopoietic stem cells. Methylcellulose-based hydrogels were fabricated and characterized based on biocompatibility with bone cells, GaAcAc loading, and thermoresponsive behavior using storage (G') and loss (G″) moduli parameters. Compared to GaAcAc solution, hydrogels loaded with GaAcAc (GaMH) were more effective in suppressing OC differentiation and function. The number and extent of bone resorption pits from ex vivo studies were markedly reduced with GaMH treatment. Mechanistic assessment of GaMH efficacy showed superiority, compared to GaAcAc solution, in downregulating the expression of key markers involved in mediating OC differentiation (such as NFAT2, cFos, TRAF6, and TRAP) as well as in bone resorption by OCs (cathepsin K or CTSK). Additional studies (in vitro and in vivo) suggested that the performance of GaMH could be ascribed to controlled release of GaAcAc and the ability to achieve prolonged bio-retention after injection in BALB/c mice, which plausibly maximized the therapeutic impact of GaAcAc. Overall, the work demonstrated, for the first time, the therapeutic efficacy of GaAcAc and the therapeutic potential of GaMH delivery systems in osteoclastic bone resorption.
Collapse
Affiliation(s)
- Pratyusha Ghanta
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
- Department of Biomedical Sciences, Kent State University, Kent, OH, 44240, USA
| | - Timothy Winschel
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Evin Hessel
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Oluyinka Oyewumi
- Department of Geological Sciences, Central Connecticut State University, New Britain, CT, 06050, USA
| | - Tori Czech
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA
| | - Moses O Oyewumi
- Advanced Drug Delivery Laboratory, Department of Pharmaceutical Sciences, College of Pharmacy, Northeast Ohio Medical University, 4209 State Route 44, Rootstown, OH, 44272, USA.
- Department of Biomedical Sciences, Kent State University, Kent, OH, 44240, USA.
| |
Collapse
|
16
|
Taira TM, Ramos-Junior ES, Melo PH, Costa-Silva CC, Alteen MG, Vocadlo DJ, Dias WB, Cunha FQ, Alves-Filho JC, Søe K, Fukada SY. HBP/O-GlcNAcylation Metabolic Axis Regulates Bone Resorption Outcome. J Dent Res 2023; 102:440-449. [PMID: 36749069 DOI: 10.1177/00220345221141043] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Osteoclasts play a key role in the regulation of bone mass and are highly active metabolically. Here we show that a metabolic reprogramming toward the hexosamine biosynthetic pathway (HBP) is required not only for osteoclast differentiation but also to determine the bone resorption mode during physiological and pathological bone remodeling. We found that pharmacological inhibition of O-GlcNAc transferase (OGT) significantly reduced protein O-GlcNAcylation and osteoclast differentiation. Accordingly, genetic deletion of OGT also inhibited osteoclast formation and downregulated critical markers related to osteoclasts differentiation and function (NFATc1, αvintegrin, cathepsin K). Indeed, cells treated with OSMI-1, an OGT inhibitor, also reduced nuclear translocation of NFATc1. Furthermore, the addition of exogenous N-acetylglucosamine (GlcNAc) strongly increased osteoclast formation and demineralization ability. Strikingly, our data show for the first time that O-GlcNAcylation facilitates an aggressive trench resorption mode in human cells. The incubation of osteoclasts with exogenous GlcNAc increases the percentage of erosion by trench while having no effect on pit resorption mode. Through time-lapse recording, we documented that osteoclasts making trenches moving across the bone surface are sensitive to GlcNAcylation. Finally, osteoclast-specific Ogt-deficient mice show increased bone density and reduced inflammation-induced bone loss during apical periodontitis model. We show that osteoclast-specific Ogt-deficient mice are less susceptible to develop bacterial-induced periapical lesion. Consistent with this, Ogt-deleted mice showed a decreased number of tartrate-resistant acid phosphatase-positive cells lining the apical periodontitis site. In summary, here we describe a hitherto undiscovered role of the HBP/O-GlcNAcylation axis tuning resorption mode and dictating bone resorption outcome.
Collapse
Affiliation(s)
- T M Taira
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, sn, 14040-903, Ribeirão Preto, Brazil
- Department of Pediatric, School of Dentistry of Ribeirão Preto, Preventive and Social Dentistry, University of São Paulo, Ribeirão Preto, Brazil
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, casa 3, 14049-900, Ribeirão Preto, Brazil
| | - E S Ramos-Junior
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, sn, 14040-903, Ribeirão Preto, Brazil
- Department of Oral Biology & Diagnostic Sciences, The Dental College of Georgia, Augusta University, Augusta, GA 30912, USA
| | - P H Melo
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Avenida Bandeirantes, 3900, 14049-900, Ribeirão Preto, Brazil
| | - C C Costa-Silva
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, sn, 14040-903, Ribeirão Preto, Brazil
| | - M G Alteen
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, B.C. V5A 1S6 Canada
| | - D J Vocadlo
- Department of Molecular Biology and Biochemistry, Simon Fraser University, 8888 University Drive, Burnaby, B.C. V5A 1S6 Canada
| | - W B Dias
- Laboratório de Glicobiologia Estrutural e Funcional, Centro de Ciências da Saúde, Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Av. Carlos Chagas Filho, 373, Rio de Janeiro, Brazil
| | - F Q Cunha
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, casa 3, 14049-900, Ribeirão Preto, Brazil
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Avenida Bandeirantes, 3900, 14049-900, Ribeirão Preto, Brazil
| | - J C Alves-Filho
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, casa 3, 14049-900, Ribeirão Preto, Brazil
- Department of Pharmacology, School of Medicine of Ribeirao Preto, University of Sao Paulo, Avenida Bandeirantes, 3900, 14049-900, Ribeirão Preto, Brazil
| | - K Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
| | - S Y Fukada
- Department of BioMolecular Sciences, School of Pharmaceutical Sciences of Ribeirao Preto, University of Sao Paulo, Avenida do Café, sn, 14040-903, Ribeirão Preto, Brazil
- Center for Research in Inflammatory Diseases, CRID, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900, casa 3, 14049-900, Ribeirão Preto, Brazil
| |
Collapse
|
17
|
Conceição F, Sousa DM, Tojal S, Lourenço C, Carvalho-Maia C, Estevão-Pereira H, Lobo J, Couto M, Rosenkilde MM, Jerónimo C, Lamghari M. The Secretome of Parental and Bone Metastatic Breast Cancer Elicits Distinct Effects in Human Osteoclast Activity after Activation of β2 Adrenergic Signaling. Biomolecules 2023; 13:biom13040622. [PMID: 37189370 DOI: 10.3390/biom13040622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 05/17/2023] Open
Abstract
The sympathetic nervous system (SNS), particularly through the β2 adrenergic receptor (β2-AR), has been linked with breast cancer (BC) and the development of metastatic BC, specifically in the bone. Nevertheless, the potential clinical benefits of exploiting β2-AR antagonists as a treatment for BC and bone loss-associated symptoms remain controversial. In this work, we show that, when compared to control individuals, the epinephrine levels in a cohort of BC patients are augmented in both earlier and late stages of the disease. Furthermore, through a combination of proteomic profiling and functional in vitro studies with human osteoclasts and osteoblasts, we demonstrate that paracrine signaling from parental BC under β2-AR activation causes a robust decrease in human osteoclast differentiation and resorption activity, which is rescued in the presence of human osteoblasts. Conversely, metastatic bone tropic BC does not display this anti-osteoclastogenic effect. In conclusion, the observed changes in the proteomic profile of BC cells under β-AR activation that take place after metastatic dissemination, together with clinical data on epinephrine levels in BC patients, provided new insights on the sympathetic control of breast cancer and its implications on osteoclastic bone resorption.
Collapse
Affiliation(s)
- Francisco Conceição
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Daniela M Sousa
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Sofia Tojal
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Catarina Lourenço
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - Carina Carvalho-Maia
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
| | - Helena Estevão-Pereira
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
| | - João Lobo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
| | - Marina Couto
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen, Denmark
| | - Carmen Jerónimo
- Cancer Biology and Epigenetics Group, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), 4200-072 Porto, Portugal
- Department of Pathology, Portuguese Oncology Institute of Porto (IPO Porto)/Porto Comprehensive Cancer Centre (Porto.CCC), 4200-072 Porto, Portugal
| | - Meriem Lamghari
- I3S-Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB-Instituto Nacional de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS-Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| |
Collapse
|
18
|
Chen ZH, Wu JJ, Guo DY, Li YY, Chen MN, Zhang ZY, Yuan ZD, Zhang KW, Chen WW, Tian F, Ye JX, Li X, Yuan FL. Physiological functions of podosomes: From structure and function to therapy implications in osteoclast biology of bone resorption. Ageing Res Rev 2023; 85:101842. [PMID: 36621647 DOI: 10.1016/j.arr.2023.101842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 12/09/2022] [Accepted: 01/03/2023] [Indexed: 01/07/2023]
Abstract
With increasing age, bone tissue undergoes significant alterations in composition, architecture, and metabolic functions, probably causing senile osteoporosis. Osteoporosis possess the vast majority of bone disease and associates with a reduction in bone mass and increased fracture risk. Bone loss is on account of the disorder in osteoblast-induced bone formation and osteoclast-induced bone resorption. As a unique bone resorptive cell type, mature bone-resorbing osteoclasts exhibit dynamic actin-based cytoskeletal structures called podosomes that participate in cell-matrix adhesions specialized in the degradation of mineralized bone matrix. Podosomes share many of the same molecular constitutions as focal adhesions, but they have a unique structural organization, with a central core abundant in F-actin and encircled by scaffolding proteins, kinases and integrins. Here, we conclude recent advancements in our knowledge of the architecture and the functions of podosomes. We also discuss the regulatory pathways in osteoclast podosomes, providing a reference for future research on the podosomes of osteoclasts and considering podosomes as a therapeutic target for inhibiting bone resorption.
Collapse
Affiliation(s)
- Zhong-Hua Chen
- Affiliated Hospital 3 of Nantong University, Nantong University, Jiangsu, China
| | - Jun-Jie Wu
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Dan-Yang Guo
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Yue-Yue Li
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Meng-Nan Chen
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Zhen-Yu Zhang
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Zheng-Dong Yuan
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Kai-Wen Zhang
- Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Wei-Wei Chen
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Fan Tian
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Jun-Xing Ye
- Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China
| | - Xia Li
- Affiliated Hospital 3 of Nantong University, Nantong University, Jiangsu, China; Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China.
| | - Feng-Lai Yuan
- Affiliated Hospital 3 of Nantong University, Nantong University, Jiangsu, China; Institute of Integrated Chinese and Western Medicine, Affiliated Hospital of Jiangnan University, Jiangsu, China.
| |
Collapse
|
19
|
Sugar transporter Slc37a2 regulates bone metabolism in mice via a tubular lysosomal network in osteoclasts. Nat Commun 2023; 14:906. [PMID: 36810735 PMCID: PMC9945426 DOI: 10.1038/s41467-023-36484-2] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 02/01/2023] [Indexed: 02/23/2023] Open
Abstract
Osteoclasts are giant bone-digesting cells that harbor specialized lysosome-related organelles termed secretory lysosomes (SLs). SLs store cathepsin K and serve as a membrane precursor to the ruffled border, the osteoclast's 'resorptive apparatus'. Yet, the molecular composition and spatiotemporal organization of SLs remains incompletely understood. Here, using organelle-resolution proteomics, we identify member a2 of the solute carrier 37 family (Slc37a2) as a SL sugar transporter. We demonstrate in mice that Slc37a2 localizes to the SL limiting membrane and that these organelles adopt a hitherto unnoticed but dynamic tubular network in living osteoclasts that is required for bone digestion. Accordingly, mice lacking Slc37a2 accrue high bone mass owing to uncoupled bone metabolism and disturbances in SL export of monosaccharide sugars, a prerequisite for SL delivery to the bone-lining osteoclast plasma membrane. Thus, Slc37a2 is a physiological component of the osteoclast's unique secretory organelle and a potential therapeutic target for metabolic bone diseases.
Collapse
|
20
|
Hansen MS, Søe K, Christensen LL, Fernandez-Guerra P, Hansen NW, Wyatt RA, Martin C, Hardy RS, Andersen TL, Olesen JB, Hartmann B, Rosenkilde MM, Kassem M, Rauch A, Gorvin CM, Frost M. GIP reduces osteoclast activity and improves osteoblast survival in primary human bone cells. Eur J Endocrinol 2023; 188:6987865. [PMID: 36747334 DOI: 10.1093/ejendo/lvac004] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 10/26/2022] [Accepted: 11/19/2022] [Indexed: 01/18/2023]
Abstract
OBJECTIVE Drugs targeting the glucose-dependent insulinotropic polypeptide (GIP) receptor (GIPR) are emerging as treatments for type-2 diabetes and obesity. GIP acutely decreases serum markers of bone resorption and transiently increases bone formation markers in short-term clinical investigations. However, it is unknown whether GIP acts directly on bone cells to mediate these effects. Using a GIPR-specific antagonist, we aimed to assess whether GIP acts directly on primary human osteoclasts and osteoblasts. METHODS Osteoclasts were differentiated from human CD14+ monocytes and osteoblasts from human bone. GIPR expression was determined using RNA-seq in primary human osteoclasts and in situ hybridization in human femoral bone. Osteoclastic resorptive activity was assessed using microscopy. GIPR signaling pathways in osteoclasts and osteoblasts were assessed using LANCE cAMP and AlphaLISA phosphorylation assays, intracellular calcium imaging and confocal microscopy. The bioenergetic profile of osteoclasts was evaluated using Seahorse XF-96. RESULTS GIPR is robustly expressed in mature human osteoclasts. GIP inhibits osteoclastogenesis, delays bone resorption, and increases osteoclast apoptosis by acting upon multiple signaling pathways (Src, cAMP, Akt, p38, Akt, NFκB) to impair nuclear translocation of nuclear factor of activated T cells-1 (NFATc1) and nuclear factor-κB (NFκB). Osteoblasts also expressed GIPR, and GIP improved osteoblast survival. Decreased bone resorption and improved osteoblast survival were also observed after GIP treatment of osteoclast-osteoblast co-cultures. Antagonizing GIPR with GIP(3-30)NH2 abolished the effects of GIP on osteoclasts and osteoblasts. CONCLUSIONS GIP inhibits bone resorption and improves survival of human osteoblasts, indicating that drugs targeting GIPR may impair bone resorption, whilst preserving bone formation.
Collapse
Affiliation(s)
- Morten S Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham B15 2TT, United Kingdom
| | - Kent Søe
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense C DK-5000, Denmark
| | - Line L Christensen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Paula Fernandez-Guerra
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Nina W Hansen
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Rachael A Wyatt
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham B15 2TT, United Kingdom
| | - Claire Martin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Rowan S Hardy
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
| | - Thomas L Andersen
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Molecular Medicine, University of Southern Denmark, Odense C DK-5000, Denmark
| | - Jacob B Olesen
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C DK-5000, Denmark
| | - Bolette Hartmann
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Mette M Rosenkilde
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen N DK-2200, Denmark
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
| | - Alexander Rauch
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Steno Diabetes Centre Odense, Odense University Hospital, Odense C DK-5000, Denmark
| | - Caroline M Gorvin
- Institute of Metabolism and Systems Research (IMSR) and Centre for Diabetes, Endocrinology and Metabolism (CEDAM), University of Birmingham, Birmingham B15 2TT, United Kingdom
- Centre for Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham B15 2TT, United Kingdom
| | - Morten Frost
- Molecular Endocrinology Laboratory (KMEB), Department of Endocrinology, Odense University Hospital, Odense C DK-5000, Denmark
- Department of Clinical Research, Faculty of Health Sciences, University of Southern Denmark, Odense C DK-5000, Denmark
- Steno Diabetes Centre Odense, Odense University Hospital, Odense C DK-5000, Denmark
| |
Collapse
|
21
|
Gomes AC, Sousa DM, Oliveira TC, Fonseca Ó, Pinto RJ, Silvério D, Fernandes AI, Moreira AC, Silva T, Teles MJ, Pereira L, Saraiva M, Lamghari M, Gomes MS. Serum amyloid A proteins reduce bone mass during mycobacterial infections. Front Immunol 2023; 14:1168607. [PMID: 37153579 PMCID: PMC10161249 DOI: 10.3389/fimmu.2023.1168607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/11/2023] [Indexed: 05/09/2023] Open
Abstract
Introduction Osteopenia has been associated to several inflammatory conditions, including mycobacterial infections. How mycobacteria cause bone loss remains elusive, but direct bone infection may not be required. Methods Genetically engineered mice and morphometric, transcriptomic, and functional analyses were used. Additionally, inflammatory mediators and bone turnover markers were measured in the serum of healthy controls, individuals with latent tuberculosis and patients with active tuberculosis. Results and discussion We found that infection with Mycobacterium avium impacts bone turnover by decreasing bone formation and increasing bone resorption, in an IFNγ- and TNFα-dependent manner. IFNγ produced during infection enhanced macrophage TNFα secretion, which in turn increased the production of serum amyloid A (SAA) 3. Saa3 expression was upregulated in the bone of both M. avium- and M. tuberculosis-infected mice and SAA1 and 2 proteins (that share a high homology with murine SAA3 protein) were increased in the serum of patients with active tuberculosis. Furthermore, the increased SAA levels seen in active tuberculosis patients correlated with altered serum bone turnover markers. Additionally, human SAA proteins impaired bone matrix deposition and increased osteoclastogenesis in vitro. Overall, we report a novel crosstalk between the cytokine-SAA network operating in macrophages and bone homeostasis. These findings contribute to a better understanding of the mechanisms of bone loss during infection and open the way to pharmacological intervention. Additionally, our data and disclose SAA proteins as potential biomarkers of bone loss during infection by mycobacteria.
Collapse
Affiliation(s)
- Ana Cordeiro Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- *Correspondence: Ana Cordeiro Gomes,
| | - Daniela Monteiro Sousa
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
| | | | - Óscar Fonseca
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Mestrado em Bioquímica Clínica, Universidade de Aveiro, , Aveiro, Portugal
| | - Ricardo J. Pinto
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Diogo Silvério
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana Isabel Fernandes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana C. Moreira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Tânia Silva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria José Teles
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- CHUSJ – Centro Hospitalar de São João, Porto, Portugal
- EPIUnit, ISPUP - Instituto de Saúde Pública da Universidade do Porto, Porto, Portugal
| | - Luísa Pereira
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IPATIMUP – Instituto de Patologia e Imunologia Molecular da Universidade do Porto, Porto, Portugal
| | - Margarida Saraiva
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- IMBC – Instituto de Biologia Molecular e Celular, Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Meriem Lamghari
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto Nacional de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Maria Salomé Gomes
- i3S – Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| |
Collapse
|
22
|
Sharma N, Weivoda MM, Søe K. Functional Heterogeneity Within Osteoclast Populations-a Critical Review of Four Key Publications that May Change the Paradigm of Osteoclasts. Curr Osteoporos Rep 2022; 20:344-355. [PMID: 35838878 DOI: 10.1007/s11914-022-00738-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2022] [Indexed: 11/03/2022]
Abstract
PURPOSE OF REVIEW In this review, we critically evaluate the literature for osteoclast heterogeneity, including heterogeneity in osteoclast behavior, which has hitherto been unstudied and has only recently come to attention. We give a critical review centered on four recent high-impact papers on this topic and aim to shed light on the elusive biology of osteoclasts and focus on the variant features of osteoclasts that diverge from the classical viewpoint. RECENT FINDINGS Osteoclasts originate from the myeloid lineage and are best known for their unique ability to resorb bone. For decades, osteoclasts have been defined simply as multinucleated cells positive for tartrate-resistant acid phosphatase activity and quantified relative to the bone perimeter or surface in histomorphometric analyses. However, several recent, high-profile studies have demonstrated the existence of heterogeneous osteoclast populations, with variable origins and functions depending on the microenvironment. This includes long-term persisting osteoclasts, inflammatory osteoclasts, recycling osteoclasts (osteomorphs), and bone resorption modes. Most of these findings have been revealed through murine studies and have helped identify new targets for human studies. These studies have also uncovered distinct sets of behavioral patterns in heterogeneous osteoclast cultures. The underlying osteoclast heterogeneity likely drives differences in bone remodeling, altering patient risk for osteoporosis and fracture. Thus, identifying the underlying key features of osteoclast heterogeneity may help in better targeting bone diseases.
Collapse
Affiliation(s)
- Neha Sharma
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark
- Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 1. Floor, 5000, Odense C, Denmark
| | | | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.
- Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.
- Department of Molecular Medicine, University of Southern Denmark, J. B. Winsløws Vej 25, 1. Floor, 5000, Odense C, Denmark.
| |
Collapse
|
23
|
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: 14] [Impact Index Per Article: 7.0] [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
|
24
|
Omi M, Mishina Y. Roles of osteoclasts in alveolar bone remodeling. Genesis 2022; 60:e23490. [PMID: 35757898 PMCID: PMC9786271 DOI: 10.1002/dvg.23490] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 05/25/2022] [Accepted: 06/09/2022] [Indexed: 12/30/2022]
Abstract
Osteoclasts are large multinucleated cells from hematopoietic origin and are responsible for bone resorption. A balance between osteoclastic bone resorption and osteoblastic bone formation is critical to maintain bone homeostasis. The alveolar bone, also called the alveolar process, is the part of the jawbone that holds the teeth and supports oral functions. It differs from other skeletal bones in several aspects: its embryonic cellular origin, the form of ossification, and the presence of teeth and periodontal tissues; hence, understanding the unique characteristic of the alveolar bone remodeling is important to maintain oral homeostasis. Excessive osteoclastic bone resorption is one of the prominent features of bone diseases in the jaw such as periodontitis. Therefore, inhibiting osteoclast formation and bone resorptive process has been the target of therapeutic intervention. Understanding the mechanisms of osteoclastic bone resorption is critical for the effective treatment of bone diseases in the jaw. In this review, we discuss basic principles of alveolar bone remodeling with a specific focus on the osteoclastic bone resorptive process and its unique functions in the alveolar bone. Lastly, we provide perspectives on osteoclast-targeted therapies and regenerative approaches associated with bone diseases in the jaw.
Collapse
Affiliation(s)
- Maiko Omi
- Department of Biologic and Materials Sciences & ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMichiganUSA
| | - Yuji Mishina
- Department of Biologic and Materials Sciences & ProsthodonticsUniversity of Michigan School of DentistryAnn ArborMichiganUSA
| |
Collapse
|
25
|
Jansen ID, van Velzen T, de Vries TJ, Szulcek R, van Loon JJWA. Real-time quantification of osteoclastic resorptive activity by electric cell-substrate impedance sensing. Front Cell Dev Biol 2022; 10:921066. [PMID: 36060806 PMCID: PMC9437204 DOI: 10.3389/fcell.2022.921066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
In several diseases, bone resorption by osteoclasts is dysregulated. Thus far, no simple technique for real-time measurement of resorption is available. Here, we introduce an impedimetric bioassay for real-time monitoring of resorption by making use of the electrical insulating properties of the resorbable substrate calcium phosphate. Different chemical stimuli were applied to (pre)osteoclasts cultured on a layer of calcium phosphate in multi-well plates containing electrodes. By this, osteoclast activity can be measured continuously over days, and the effects of stimulating or inhibiting factors can be quantified. When cells were cultured in the presence of an inflammatory factor such as IL-1β, the resorptive activity started earlier. The measured decline in resistance was higher at culture day 5 than at cultures with M-CSF or M-CSF + RANKL (M-CSF norm. Resistance = 1, M-CSF + RANKL = 0.7, M-CSF + RANKL + IL-1β = 0.5). However, at day 11, this difference had nearly disappeared. Likewise, bisphosphonates were shown to inhibit osteoclastic activity. Our findings illustrate the importance of real-time monitoring; wherefore, this method has high potential not only for the study of osteoclast resorptive activity in the context of osteoclast function and diseases but also could find application in high-throughput drug-testing studies.
Collapse
Affiliation(s)
- Ineke D.C. Jansen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Thijs van Velzen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
| | - Robert Szulcek
- Department of Pulmonary Diseases, Amsterdam UMC, VU University Medical Center, Amsterdam Cardiovascular Sciences (ACS), Amsterdam, Netherlands
- Laboratory of in vitro Modeling Systems of Pulmonary and Thrombotic Diseases, Institute of Physiology, Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Heart Center Berlin, Berlin, Germany
| | - Jack J. W. A. van Loon
- Life Support and Physical Sciences Section (TEC-MMG), European Space Agency—European Space Research and Technology Centre (ESA-ESTEC), Noordwijk, Netherlands
- DESC (Dutch Experiment Support Center), Amsterdam University Medical Center Location VUmc, Amsterdam, Netherlands
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Department of Oral and Maxillofacial Surgery/Pathology, Amsterdam University Medical Center Amsterdam Bone Center (ABC), Amsterdam, Netherlands
- *Correspondence: Jack J. W. A. van Loon,
| |
Collapse
|
26
|
Swanson WB, Durdan M, Eberle M, Woodbury S, Mauser A, Gregory J, Zhang B, Niemann D, Herremans J, Ma PX, Lahann J, Weivoda M, Mishina Y, Greineder CF. A library of Rhodamine6G-based pH-sensitive fluorescent probes with versatile in vivo and in vitro applications. RSC Chem Biol 2022; 3:748-764. [PMID: 35755193 PMCID: PMC9175114 DOI: 10.1039/d2cb00030j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Accepted: 04/24/2022] [Indexed: 01/11/2023] Open
Abstract
Acidic pH is critical to the function of the gastrointestinal system, bone-resorbing osteoclasts, and the endolysosomal compartment of nearly every cell in the body. Non-invasive, real-time fluorescence imaging of acidic microenvironments represents a powerful tool for understanding normal cellular biology, defining mechanisms of disease, and monitoring for therapeutic response. While commercially available pH-sensitive fluorescent probes exist, several limitations hinder their widespread use and potential for biologic application. To address this need, we developed a novel library of pH-sensitive probes based on the highly photostable and water-soluble fluorescent molecule, Rhodamine 6G. We demonstrate versatility in terms of both pH sensitivity (i.e., pK a) and chemical functionality, allowing conjugation to small molecules, proteins, nanoparticles, and regenerative biomaterial scaffold matrices. Furthermore, we show preserved pH-sensitive fluorescence following a variety of forms of covalent functionalization and demonstrate three potential applications, both in vitro and in vivo, for intracellular and extracellular pH sensing. Finally, we develop a computation approach for predicting the pH sensitivity of R6G derivatives, which could be used to expand our library and generate probes with novel properties.
Collapse
Affiliation(s)
- W Benton Swanson
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
| | - Margaret Durdan
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Cell and Molecular Biology Program, Medical School, University of Michigan Ann Arbor MI USA
| | - Miranda Eberle
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan Ann Arbor MI USA
| | - Seth Woodbury
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan Ann Arbor MI USA
| | - Ava Mauser
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Biomedical Engineering, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
| | - Jason Gregory
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Chemical Engineering, College of Engineering, University of Michigan Ann Arbor MI USA
| | - Boya Zhang
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Pharmacology, Medical School, University of Michigan Ann Arbor MI USA
| | - David Niemann
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan Ann Arbor MI USA
- Department of Chemical Engineering, College of Engineering, University of Michigan Ann Arbor MI USA
| | - Jacob Herremans
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
- Department of Chemistry, College of Literature, Science and the Arts, University of Michigan Ann Arbor MI USA
| | - Peter X Ma
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
- Department of Biomedical Engineering, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Materials Science and Engineering, College of Engineering, University of Michigan Ann Arbor MI USA
- Macromolecular Science and Engineering Center, College of Engineering, University of Michigan Ann Arbor MI USA
| | - Joerg Lahann
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Biomedical Engineering, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Chemical Engineering, College of Engineering, University of Michigan Ann Arbor MI USA
- Department of Materials Science and Engineering, College of Engineering, University of Michigan Ann Arbor MI USA
- Macromolecular Science and Engineering Center, College of Engineering, University of Michigan Ann Arbor MI USA
| | - Megan Weivoda
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Pharmacology, Medical School, University of Michigan Ann Arbor MI USA
- Department of Periodontics and Oral Medicine, School of Dentistry, University of Michigan Ann Arbor MI USA
| | - Yuji Mishina
- Department of Biologic and Materials Science, School of Dentistry, University of Michigan 1011 North University Avenue Ann Arbor MI 48109 USA
| | - Colin F Greineder
- Biointerfaces Institute, College of Engineering and Medical School, University of Michigan Ann Arbor MI USA
- Department of Pharmacology, Medical School, University of Michigan Ann Arbor MI USA
- Department of Emergency Medicine, Medical School, University of Michigan NCRC 2800 Plymouth Road, Bldg #26 Ann Arbor MI 48109 USA
| |
Collapse
|
27
|
Malek G, Richard H, Beauchamp G, Laverty S. An in vitro model for discovery of osteoclast specific biomarkers towards identification of racehorses at risk for catastrophic fractures. Equine Vet J 2022; 55:534-550. [PMID: 35616632 DOI: 10.1111/evj.13600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 05/12/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND Focal bone microcracks with osteoclast recruitment and bone lysis, may reduce fracture resistance in racehorses. As current imaging does not detect all horses at risk for fracture, the discovery of novel serum biomarkers of bone resorption or osteoclast activity could potentially address this unmet clinical need. The biology of equine osteoclasts on their natural substrate, equine bone, has never been studied in vitro and may permit identification of specific biomarkers of their activity. OBJECTIVES 1) Establish osteoclast cultures on equine bone, 2) Measure biomarkers (tartrate resistant acid phosphatase isoform 5b (TRACP-5b) and C-terminal telopeptide of type I collagen (CTX-I)) in vitro and 3) Study the effects of inflammation. STUDY DESIGN In vitro experiments. METHODS Haematopoietic stem cells, from 5 equine sternal bone marrow aspirates, were differentiated into osteoclasts and cultured either alone or on equine bone slices, with or without pro-inflammatory stimulus (IL-1β or LPS). CTX-I and TRACP-5b were immunoassayed in the media. Osteoclast numbers and bone resorption area were assessed. RESULTS TRACP-5b increased over time without bone (p < 0.0001) and correlated with osteoclast number (r = 0.63, p < 0.001). CTX-I and TRACP-5b increased with time for cultures with bone (p = 0.002; p = 0.02 respectively), correlated with each other (r = 0.64, p < 0.002) and correlated with bone resorption (r = 0.85, p < 0.001; r = 0.82, p < 0.001 respectively). Inflammation had no measurable effects. MAIN LIMITATIONS Specimen numbers limited. CONCLUSIONS Equine osteoclasts were successfully cultured on equine bone slices and their bone resorption quantified. TRACP-5b was shown to be a biomarker of equine osteoclast number and bone resorption for the first time; CTX-I was also confirmed to be a biomarker of equine bone resorption in vitro. This robust equine specific in vitro assay will help the study of osteoclast biology.
Collapse
Affiliation(s)
- Gwladys Malek
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC, Canada
| | - Hélène Richard
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC, Canada
| | - Guy Beauchamp
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC, Canada
| | - Sheila Laverty
- Comparative Orthopaedic Research Laboratory, Department of Clinical Sciences, Faculty of Veterinary Medicine, University of Montreal, 3200 Sicotte, St-Hyacinthe, QC, Canada
| |
Collapse
|
28
|
Comninos AN, Hansen MS, Courtney A, Choudhury S, Yang L, Mills EG, Phylactou M, Busbridge M, Khir M, Thaventhiran T, Bech P, Tan T, Abbara A, Frost M, Dhillo WS. Acute Effects of Kisspeptin Administration on Bone Metabolism in Healthy Men. J Clin Endocrinol Metab 2022; 107:1529-1540. [PMID: 35244717 PMCID: PMC9113799 DOI: 10.1210/clinem/dgac117] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Indexed: 12/23/2022]
Abstract
CONTEXT Osteoporosis results from disturbances in bone formation and resorption. Recent nonhuman data suggest that the reproductive hormone kisspeptin directly stimulates osteoblast differentiation in vitro and thus could have clinical therapeutic potential. However, the effects of kisspeptin on human bone metabolism are currently unknown. OBJECTIVE To assess the effects of kisspeptin on human bone metabolism in vitro and in vivo. METHODS In vitro study: of Mono- and cocultures of human osteoblasts and osteoclasts treated with kisspeptin. Clinical study: Randomized, placebo-controlled, double-blind, 2-way crossover clinical study in 26 men investigating the effects of acute kisspeptin administration (90 minutes) on human bone metabolism, with blood sampling every 30 minutes to +90 minutes. Cells for the in vitro study were from 12 male blood donors and 8 patients undergoing hip replacement surgery. Twenty-six healthy eugonadal men (age 26.8 ± 5.8 years) were included in the clinical study. The intervention was Kisspeptin (vs placebo) administration. The main outcome measures were changes in bone parameters and turnover markers. RESULTS Incubation with kisspeptin in vitro increased alkaline phosphatase levels in human bone marrow mesenchymal stem cells by 41.1% (P = .0022), and robustly inhibited osteoclastic resorptive activity by up to 53.4% (P < .0001), in a dose-dependent manner. Kisspeptin administration to healthy men increased osteoblast activity, as evidenced by a 20.3% maximal increase in total osteocalcin (P = .021) and 24.3% maximal increase in carboxylated osteocalcin levels (P = .014). CONCLUSION Collectively, these data provide the first human evidence that kisspeptin promotes osteogenic differentiation of osteoblast progenitors and inhibits bone resorption in vitro. Furthermore, kisspeptin acutely increases the bone formation marker osteocalcin but not resorption markers in healthy men, independent of downstream sex steroid levels. Kisspeptin could therefore have clinical therapeutic application in the treatment of osteoporosis.
Collapse
Affiliation(s)
- Alexander N Comninos
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
- Endocrine Bone Unit, Imperial College Healthcare NHS Trust, London, UK
| | - Morten S Hansen
- KMEB Molecular Endocrinology Laboratory, Department of Endocrinology, Odense University Hospital, Denmark
- Department of Clinical Research, University of Southern Denmark, Denmark
| | - Alan Courtney
- Department of Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, UK
| | - Sirazum Choudhury
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, UK
| | - Lisa Yang
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Edouard G Mills
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Maria Phylactou
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Mark Busbridge
- Department of Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, UK
| | - Muaza Khir
- Department of Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, UK
| | - Thilipan Thaventhiran
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Paul Bech
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, UK
| | - Tricia Tan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Clinical Biochemistry, Imperial College Healthcare NHS Trust, London, UK
| | - Ali Abbara
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| | - Morten Frost
- KMEB Molecular Endocrinology Laboratory, Department of Endocrinology, Odense University Hospital, Denmark
- Department of Clinical Research, University of Southern Denmark, Denmark
- Steno Diabetes Centre, Odense University Hospital, Denmark
| | - Waljit S Dhillo
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
- Department of Endocrinology, Imperial College Healthcare NHS Trust, London, UK
| |
Collapse
|
29
|
Parsegian K, Randall D, Curtis M, Ioannidou E. Association between periodontitis and chronic kidney disease. Periodontol 2000 2022; 89:114-124. [PMID: 35244955 DOI: 10.1111/prd.12431] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Periodontitis and chronic kidney disease are chronic conditions with high community prevalence across the world. Patients with chronic kidney disease have been noted to have a high burden of periodontitis, and several shared risk factors have been associated with the prevalence and severity of both conditions. However, the precise relationship between the two conditions, and the extent to which each may contribute to the development of the other, remains a matter of debate. The goals of the present work were to: (a) provide the most current and relevant literature overview of the association between periodontitis and chronic kidney disease; (b) explore mechanisms underlying this association; and (c) determine if evidence exists for an independent association between these conditions. We also assessed whether improved oral hygiene and periodontal treatment could reduce the risk of developing chronic kidney disease and, if so, what protocols these strategies involve. Finally, we aimed to reveal gaps in our current knowledge to delineate the directions of future research. Although the exact relationship between these two conditions has not yet been defined, we highlight the importance of the interprofessional interaction between dental practitioners and the nephrology team and the importance of oral health assessment in the management of chronic kidney disease.
Collapse
Affiliation(s)
- Karo Parsegian
- Department of Surgical Dentistry, Division of Periodontics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - David Randall
- William Harvey Research Institute, Charterhouse Square Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Mike Curtis
- Faculty of Dentistry, Oral & Craniofacial Sciences, Guy's Hospital, London, UK
| | - Effie Ioannidou
- Department of Oral Health and Diagnostic Sciences, Division of Periodontology, Dental Clinical Research Center, School of Dental Medicine, University of Connecticut Health, Farmington, Connecticut, USA
| |
Collapse
|
30
|
A metastasis-on-a-chip approach to explore the sympathetic modulation of breast cancer bone metastasis. Mater Today Bio 2022; 13:100219. [PMID: 35243294 PMCID: PMC8857466 DOI: 10.1016/j.mtbio.2022.100219] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/10/2022] [Accepted: 02/12/2022] [Indexed: 01/09/2023]
Abstract
Organ-on-a-chip models have emerged as a powerful tool to model cancer metastasis and to decipher specific crosstalk between cancer cells and relevant regulators of this particular niche. Recently, the sympathetic nervous system (SNS) was proposed as an important modulator of breast cancer bone metastasis. However, epidemiological studies concerning the benefits of the SNS targeting drugs on breast cancer survival and recurrence remain controversial. Thus, the role of SNS signaling over bone metastatic cancer cellular processes still requires further clarification. Herein, we present a novel humanized organ-on-a-chip model recapitulating neuro-breast cancer crosstalk in a bone metastatic context. We developed and validated an innovative three-dimensional printing based multi-compartment microfluidic platform, allowing both selective and dynamic multicellular paracrine signaling between sympathetic neurons, bone tropic breast cancer cells and osteoclasts. The selective multicellular crosstalk in combination with biochemical, microscopic and proteomic profiling show that synergistic paracrine signaling from sympathetic neurons and osteoclasts increase breast cancer aggressiveness demonstrated by augmented levels of pro-inflammatory cytokines (e.g. interleukin-6 and macrophage inflammatory protein 1α). Overall, this work introduced a novel and versatile platform that could potentially be used to unravel new mechanisms involved in intracellular communication at the bone metastatic niche.
Collapse
|
31
|
Rauner M, Foessl I, Formosa MM, Kague E, Prijatelj V, Lopez NA, Banerjee B, Bergen D, Busse B, Calado Â, Douni E, Gabet Y, Giralt NG, Grinberg D, Lovsin NM, Solan XN, Ostanek B, Pavlos NJ, Rivadeneira F, Soldatovic I, van de Peppel J, van der Eerden B, van Hul W, Balcells S, Marc J, Reppe S, Søe K, Karasik D. Perspective of the GEMSTONE Consortium on Current and Future Approaches to Functional Validation for Skeletal Genetic Disease Using Cellular, Molecular and Animal-Modeling Techniques. Front Endocrinol (Lausanne) 2021; 12:731217. [PMID: 34938269 PMCID: PMC8686830 DOI: 10.3389/fendo.2021.731217] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 09/30/2021] [Indexed: 12/26/2022] Open
Abstract
The availability of large human datasets for genome-wide association studies (GWAS) and the advancement of sequencing technologies have boosted the identification of genetic variants in complex and rare diseases in the skeletal field. Yet, interpreting results from human association studies remains a challenge. To bridge the gap between genetic association and causality, a systematic functional investigation is necessary. Multiple unknowns exist for putative causal genes, including cellular localization of the molecular function. Intermediate traits ("endophenotypes"), e.g. molecular quantitative trait loci (molQTLs), are needed to identify mechanisms of underlying associations. Furthermore, index variants often reside in non-coding regions of the genome, therefore challenging for interpretation. Knowledge of non-coding variance (e.g. ncRNAs), repetitive sequences, and regulatory interactions between enhancers and their target genes is central for understanding causal genes in skeletal conditions. Animal models with deep skeletal phenotyping and cell culture models have already facilitated fine mapping of some association signals, elucidated gene mechanisms, and revealed disease-relevant biology. However, to accelerate research towards bridging the current gap between association and causality in skeletal diseases, alternative in vivo platforms need to be used and developed in parallel with the current -omics and traditional in vivo resources. Therefore, we argue that as a field we need to establish resource-sharing standards to collectively address complex research questions. These standards will promote data integration from various -omics technologies and functional dissection of human complex traits. In this mission statement, we review the current available resources and as a group propose a consensus to facilitate resource sharing using existing and future resources. Such coordination efforts will maximize the acquisition of knowledge from different approaches and thus reduce redundancy and duplication of resources. These measures will help to understand the pathogenesis of osteoporosis and other skeletal diseases towards defining new and more efficient therapeutic targets.
Collapse
Affiliation(s)
- Martina Rauner
- Department of Medicine III, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
- University Hospital Carl Gustav Carus, Dresden, Germany
| | - Ines Foessl
- Department of Internal Medicine, Division of Endocrinology and Diabetology, Endocrine Lab Platform, Medical University of Graz, Graz, Austria
| | - Melissa M. Formosa
- Department of Applied Biomedical Science, Faculty of Health Sciences, University of Malta, Msida, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Msida, Malta
| | - Erika Kague
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
| | - Vid Prijatelj
- Department of Oral and Maxillofacial Surgery, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- The Generation R Study, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Nerea Alonso Lopez
- Rheumatology and Bone Disease Unit, CGEM, Institute of Genetics and Cancer (IGC), Edinburgh, United Kingdom
| | - Bodhisattwa Banerjee
- Musculoskeletal Genetics Laboratory, Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Dylan Bergen
- School of Physiology, Pharmacology, and Neuroscience, Faculty of Life Sciences, University of Bristol, Bristol, United Kingdom
- Musculoskeletal Research Unit, Translational Health Sciences, Bristol Medical School, Faculty of Health Sciences, University of Bristol, Bristol, United Kingdom
| | - Björn Busse
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ângelo Calado
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Centro Académico de Medicina de Lisboa, Lisbon, Portugal
| | - Eleni Douni
- Department of Biotechnology, Agricultural University of Athens, Athens, Greece
- Institute for Bioinnovation, B.S.R.C. “Alexander Fleming”, Vari, Greece
| | - Yankel Gabet
- Department of Anatomy & Anthropology, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Natalia García Giralt
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Daniel Grinberg
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - Nika M. Lovsin
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Xavier Nogues Solan
- Musculoskeletal Research Group, IMIM (Hospital del Mar Medical Research Institute), Centro de Investigación Biomédica en Red en Fragilidad y Envejecimiento Saludable (CIBERFES), ISCIII, Barcelona, Spain
| | - Barbara Ostanek
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Nathan J. Pavlos
- Bone Biology & Disease Laboratory, School of Biomedical Sciences, The University of Western Australia, Nedlands, WA, Australia
| | | | - Ivan Soldatovic
- Institute of Medical Statistics and Informatic, Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Jeroen van de Peppel
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Bram van der Eerden
- Department of Internal Medicine, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Wim van Hul
- Department of Medical Genetics, University of Antwerp, Antwerp, Belgium
| | - Susanna Balcells
- Department of Genetics, Microbiology and Statistics, Faculty of Biology, Universitat de Barcelona, CIBERER, IBUB, IRSJD, Barcelona, Spain
| | - Janja Marc
- Department of Clinical Biochemistry, Faculty of Pharmacy, University of Ljubljana, Ljubljana, Slovenia
| | - Sjur Reppe
- Unger-Vetlesen Institute, Lovisenberg Diaconal Hospital, Oslo, Norway
- Department of Plastic and Reconstructive Surgery, Oslo University Hospital, Oslo, Norway
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Kent Søe
- Clinical Cell Biology, 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
| | - David Karasik
- Azrieli Faculty of Medicine, Bar-Ilan University, Ramat Gan, Israel
- Marcus Research Institute, Hebrew SeniorLife, Boston, MA, United States
| |
Collapse
|
32
|
Salamanna F, Maglio M, Borsari V, Landini MP, Fini M. Blood factors as biomarkers in osteoporosis: points from the COVID-19 era. Trends Endocrinol Metab 2021; 32:672-679. [PMID: 34246532 PMCID: PMC8261630 DOI: 10.1016/j.tem.2021.05.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 05/18/2021] [Accepted: 05/24/2021] [Indexed: 11/20/2022]
Abstract
The restrictions adopted during the coronavirus disease 2019 (COVID-19) pandemic limiting direct medical consultations and access to healthcare centers reduced the participation of patients with chronic diseases, such as osteoporosis (OP), in screening and monitoring programs. This highlighted the need for new screening diagnostic tools that are clinically effective, but require minimal technical and time commitments, to stratify populations and identify who is more at risk for OP and related complications. This paper provides an overview of the potential use of blood-related factors, such as platelet (PLT)- and monocyte-related factors, as biomarkers able to quickly screen, detect, and monitor OP in both sexes. Such biomarkers might be of key importance not only during the COVID-19 pandemic but also, even more importantly, during periods of better global health stability.
Collapse
Affiliation(s)
- Francesca Salamanna
- IRCCS Istituto Ortopedico Rizzoli, Complex Structure of Surgical Sciences and Technologies, via di Barbiano 1/10, Bologna 40136, Italy
| | - Melania Maglio
- IRCCS Istituto Ortopedico Rizzoli, Complex Structure of Surgical Sciences and Technologies, via di Barbiano 1/10, Bologna 40136, Italy.
| | - Veronica Borsari
- IRCCS Istituto Ortopedico Rizzoli, Complex Structure of Surgical Sciences and Technologies, via di Barbiano 1/10, Bologna 40136, Italy
| | - Maria Paola Landini
- IRCCS Istituto Ortopedico Rizzoli, Scientific Direction, via di Barbiano 1/10, Bologna 40136, Italy
| | - Milena Fini
- IRCCS Istituto Ortopedico Rizzoli, Complex Structure of Surgical Sciences and Technologies, via di Barbiano 1/10, Bologna 40136, Italy
| |
Collapse
|
33
|
Rössler U, Hennig AF, Stelzer N, Bose S, Kopp J, Søe K, Cyganek L, Zifarelli G, Ali S, von der Hagen M, Strässler ET, Hahn G, Pusch M, Stauber T, Izsvák Z, Gossen M, Stachelscheid H, Kornak U. Efficient generation of osteoclasts from human induced pluripotent stem cells and functional investigations of lethal CLCN7-related osteopetrosis. J Bone Miner Res 2021; 36:1621-1635. [PMID: 33905594 DOI: 10.1002/jbmr.4322] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 03/26/2021] [Accepted: 04/21/2021] [Indexed: 12/18/2022]
Abstract
Human induced pluripotent stem cells (hiPSCs) hold great potential for modeling human diseases and the development of innovative therapeutic approaches. Here, we report on a novel, simplified differentiation method for forming functional osteoclasts from hiPSCs. The three-step protocol starts with embryoid body formation, followed by hematopoietic specification, and finally osteoclast differentiation. We observed continuous production of monocyte-like cells over a period of up to 9 weeks, generating sufficient material for several osteoclast differentiations. The analysis of stage-specific gene and surface marker expression proved mesodermal priming, the presence of monocyte-like cells, and of terminally differentiated multinucleated osteoclasts, able to form resorption pits and trenches on bone and dentine in vitro. In comparison to peripheral blood mononuclear cell (PBMC)-derived osteoclasts hiPSC-derived osteoclasts were larger and contained a higher number of nuclei. Detailed functional studies on the resorption behavior of hiPSC-osteoclasts indicated a trend towards forming more trenches than pits and an increase in pseudoresorption. We used hiPSCs from an autosomal recessive osteopetrosis (ARO) patient (BIHi002-A, ARO hiPSCs) with compound heterozygous missense mutations p.(G292E) and p.(R403Q) in CLCN7, coding for the Cl- /H+ -exchanger ClC-7, for functional investigations. The patient's leading clinical feature was a brain malformation due to defective neuronal migration. Mutant ClC-7 displayed residual expression and retained lysosomal co-localization with OSTM1, the gene coding for the osteopetrosis-associated transmembrane protein 1, but only ClC-7 harboring the mutation p.(R403Q) gave strongly reduced ion currents. An increased autophagic flux in spite of unchanged lysosomal pH was evident in undifferentiated ARO hiPSCs. ARO hiPSC-derived osteoclasts showed an increased size compared to hiPSCs of healthy donors. They were not able to resorb bone, underlining a loss-of-function effect of the mutations. In summary, we developed a highly reproducible, straightforward hiPSC-osteoclast differentiation protocol. We demonstrated that osteoclasts differentiated from ARO hiPSCs can be used as a disease model for ARO and potentially also other osteoclast-related diseases. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Uta Rössler
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), Berlin, Germany
| | - Anna Floriane Hennig
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Freie Universität Berlin, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
| | - Nina Stelzer
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Shroddha Bose
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany
| | - Johannes Kopp
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Freie Universität Berlin, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense C, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense M, Denmark.,Department of Molecular Medicine, University of Southern Denmark, Odense M, Denmark
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, Göttingen, Germany.,German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, Göttingen, Germany
| | | | - Salaheddine Ali
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| | - Maja von der Hagen
- Abteilung Neuropädiatrie, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Elisabeth Tamara Strässler
- Department of Cardiology, Charité - Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany.,German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Gabriele Hahn
- Institut und Poliklinik für Radiologische Diagnostik, Medizinische Fakultät Carl Gustav Carus Technische Universität Dresden, Dresden, Germany
| | | | - Tobias Stauber
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.,Department of Human Medicine, and Institute for Molecular Medicine, MSH Medical School Hamburg, Hamburg, Germany
| | - Zsuzsanna Izsvák
- Max-Delbrück-Center for Molecular Medicine (MDC), Helmholtz Association, Berlin, Germany
| | - Manfred Gossen
- Berlin-Brandenburg Center for Regenerative Therapies, Charité Virchow Campus, Berlin, Germany.,Institute of Active Polymers, Helmholtz-Zentrum Hereon, Teltow, Germany
| | - Harald Stachelscheid
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Berlin Institute of Health (BIH), BIH Stem Cell Core Facility, Berlin, Germany
| | - Uwe Kornak
- BIH Center for Regenerative Therapies (BCRT), Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute for Medical Genetics and Human Genetics, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany.,Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany.,Max Planck Institute for Molecular Genetics, Berlin, Germany
| |
Collapse
|
34
|
Lee SH, Ihn HJ, Park EK, Kim JE. S100 Calcium-Binding Protein P Secreted from Megakaryocytes Promotes Osteoclast Maturation. Int J Mol Sci 2021; 22:ijms22116129. [PMID: 34200172 PMCID: PMC8201154 DOI: 10.3390/ijms22116129] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Revised: 05/31/2021] [Accepted: 06/02/2021] [Indexed: 12/26/2022] Open
Abstract
Megakaryocytes (MKs) differentiate from hematopoietic stem cells and produce platelets at the final stage of differentiation. MKs directly interact with bone cells during bone remodeling. However, whether MKs are involved in regulating bone metabolism through indirect regulatory effects on bone cells is unclear. Here, we observed increased osteoclast differentiation of bone marrow-derived macrophages (BMMs) cultured in MK-cultured conditioned medium (MK CM), suggesting that this medium contains factors secreted from MKs that affect osteoclastogenesis. To identify the MK-secreted factor, DNA microarray analysis of the human leukemia cell line K562 and MKs was performed, and S100 calcium-binding protein P (S100P) was selected as a candidate gene affecting osteoclast differentiation. S100P was more highly expressed in MKs than in K562 cells, and showed higher levels in MK CM than in K562-cultured conditioned medium. In BMMs cultured in the presence of recombinant human S100P protein, osteoclast differentiation was promoted and marker gene expression was increased. The resorption area was significantly larger in S100P protein-treated osteoclasts, demonstrating enhanced resorption activity. Overall, S100P secreted from MKs promotes osteoclast differentiation and resorption activity, suggesting that MKs indirectly regulate osteoclast differentiation and activity through the paracrine action of S100P.
Collapse
Affiliation(s)
- Seung-Hoon Lee
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea;
| | - Hye Jung Ihn
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea;
| | - Eui Kyun Park
- Department of Oral Pathology and Regenerative Medicine, School of Dentistry, IHBR, Kyungpook National University, Daegu 41944, Korea;
| | - Jung-Eun Kim
- Department of Molecular Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Korea;
- BK21 Four KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, Kyungpook National University, Daegu 41944, Korea
- Cell and Matrix Research Institute, Kyungpook National University, Daegu 41944, Korea;
- Correspondence: ; Tel.: +82-53-420-4949
| |
Collapse
|
35
|
Richard ET, Morinaga K, Zheng Y, Sundberg O, Hokugo A, Hui K, Zhou Y, Sasaki H, Kashemirov BA, Nishimura I, McKenna CE. Design and Synthesis of Cathepsin-K-Activated Osteoadsorptive Fluorogenic Sentinel (OFS) Probes for Detecting Early Osteoclastic Bone Resorption in a Multiple Myeloma Mouse Model. Bioconjug Chem 2021; 32:916-927. [PMID: 33956423 PMCID: PMC8137654 DOI: 10.1021/acs.bioconjchem.1c00036] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
We describe the design and synthesis of OFS-1, an Osteoadsorptive Fluorogenic Sentinel imaging probe that is adsorbed by hydroxyapatite (HAp) and bone mineral surfaces, where it generates an external fluorescent signal in response to osteoclast-secreted cathepsin K (Ctsk). The probe consists of a bone-anchoring bisphosphonate moiety connected to a Förster resonance energy transfer (FRET) internally quenched fluorescent (IQF) dye pair, linked by a Ctsk peptide substrate, GHPGGPQG. Key structural features contributing to the effectiveness of OFS-1 were defined by structure-activity relationship (SAR) and modeling studies comparing OFS-1 with two cognates, OFS-2 and OFS-3. In solution or when preadsorbed on HAp, OFS-1 exhibited strong fluorescence when exposed to Ctsk (2.5-20 nM). Time-lapse photomicrographs obtained after seeding human osteoclasts onto HAp-coated well plates containing preadsorbed OFS-1 revealed bright fluorescence at the periphery of resorbing cells. OFS-1 administered systemically detected early osteolysis colocalized with orthotopic engraftment of RPMI-8226-Luc human multiple myeloma cells at a metastatic skeletal site in a humanized mouse model. OFS-1 is thus a promising new imaging tool for detecting abnormal bone resorption.
Collapse
Affiliation(s)
- Eric T. Richard
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
- Center for Craniofacial Molecular Biology, Herman Ostrow School of Dentistry, University of Southern California, Los Angeles, California, 90033, USA
| | - Kenzo Morinaga
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, California, 90095, USA
- Department of Oral Rehabilitation, Section of Oral Implantology, Fukuoka Dental College, Fukuoka, Japan
| | - Yiying Zheng
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
| | - Oskar Sundberg
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
| | - Akishige Hokugo
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, California, 90095, USA
- Division of Plastic and Reconstructive Surgery, David Geffen School of Medicine, UCLA, Los Angeles, California, 90095, USA
| | - Kimberly Hui
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
| | - Yipin Zhou
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
| | - Hodaka Sasaki
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, California, 90095, USA
- Department of Oral and Maxillofacial Implantology, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Boris A. Kashemirov
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
| | - Ichiro Nishimura
- Weintraub Center for Reconstructive Biotechnology, UCLA School of Dentistry, Los Angeles, California, 90095, USA
| | - Charles E. McKenna
- Department of Chemistry, University of Southern California, Los Angeles, California, 90089, USA
| |
Collapse
|
36
|
Simon P, Pompe W, Bobeth M, Worch H, Kniep R, Formanek P, Hild A, Wenisch S, Sturm E. Podosome-Driven Defect Development in Lamellar Bone under the Conditions of Senile Osteoporosis Observed at the Nanometer Scale. ACS Biomater Sci Eng 2021; 7:2255-2267. [PMID: 33938726 PMCID: PMC8290401 DOI: 10.1021/acsbiomaterials.0c01493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The degradation mechanism of human trabecular bone harvested from the central part of the femoral head of a patient with a fragility fracture of the femoral neck under conditions of senile osteoporosis was investigated by high-resolution electron microscopy. As evidenced by light microscopy, there is a disturbance of bone metabolism leading to severe and irreparable damages to the bone structure. These defects are evoked by osteoclasts and thus podosome activity. Podosomes create typical pit marks and holes of about 300-400 nm in diameter on the bone surface. Detailed analysis of the stress field caused by the podosomes in the extracellular bone matrix was performed. The calculations yielded maximum stress in the range of few megapascals resulting in formation of microcracks around the podosomes. Disintegration of hydroxyapatite and free lying collagen fibrils were observed at the edges of the plywood structure of the bone lamella. At the ultimate state, the disintegration of the mineralized collagen fibrils to a gelatinous matrix comes along with a delamination of the apatite nanoplatelets resulting in a brittle, porous bone structure. The nanoplatelets aggregate to big hydroxyapatite plates with a size of up to 10 x 20 μm2. The enhanced plate growth can be explained by the interaction of two mechanisms in the ruffled border zone: the accumulation of delaminated hydroxyapatite nanoplatelets near clusters of podosomes and the accelerated nucleation and random growth of HAP nanoplatelets due to a nonsufficient concentration of process-directing carboxylated osteocalcin cOC.
Collapse
Affiliation(s)
- Paul Simon
- Max-Planck-Institut für Chemische Physik fester Stoffe, Nöthnitzer Str. 40, 01187 Dresden, Germany
| | - Wolfgang Pompe
- Technical University of Dresden, Institute of Materials Science, 01069 Dresden, Germany
| | - Manfred Bobeth
- Technical University of Dresden, Institute of Materials Science, 01069 Dresden, Germany
| | - Hartmut Worch
- Technical University of Dresden, Institute of Materials Science, 01069 Dresden, Germany
| | - Rüdiger Kniep
- Max-Planck-Institut für Chemische Physik fester Stoffe, Nöthnitzer Str. 40, 01187 Dresden, Germany
| | - Petr Formanek
- Leibniz-Institut für Polymerforschung Dresden e.V., Hohe Straße 6, 01069 Dresden, Germany
| | - Anne Hild
- Clinical Anatomy, Clinic of Small Animals, Justus-Liebig-University, 35385 Giessen, Germany
| | - Sabine Wenisch
- Clinical Anatomy, Clinic of Small Animals, Justus-Liebig-University, 35385 Giessen, Germany
| | - Elena Sturm
- Max-Planck-Institut für Chemische Physik fester Stoffe, Nöthnitzer Str. 40, 01187 Dresden, Germany.,University of Konstanz, Physical Chemistry, POB 714, D-78457 Konstanz, Germany
| |
Collapse
|
37
|
Chen S, Wang Q, Eltit F, Guo Y, Cox M, Wang R. An Ammonia-Induced Calcium Phosphate Nanostructure: A Potential Assay for Studying Osteoporosis and Bone Metastasis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:17207-17219. [PMID: 33845570 DOI: 10.1021/acsami.1c00495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Osteoclastic resorption of bones plays a central role in both osteoporosis and bone metastasis. A reliable in vitro assay that simulates osteoclastic resorption in vivo would significantly speed up the process of developing effective therapeutic solutions for those diseases. Here, we reported the development of a novel and robust nanostructured calcium phosphate coating with unique functions on the track-etched porous membrane by using an ammonia-induced mineralization (AiM) technique. The calcium phosphate coating uniformly covers one side of the PET membrane, enabling testing for osteoclastic resorption. The track-etched pores in the PET membrane allow calcium phosphate mineral pins to grow inside, which, on the one hand, enhances coating integration with a membrane substrate and, on the other hand, provides diffusion channels for delivering drugs from the lower chamber of a double-chamber cell culture system. The applications of the processed calcium phosphate coating were first demonstrated as a drug screening device by using alendronate, a widely used drug for osteoporosis. It was confirmed that the delivery of alendronate significantly decreased both the number of monocyte-differentiated osteoclasts and coating resorption. To demonstrate the application in studying bone metastasis, we delivered a PC3 prostate cancer-conditioned medium and confirmed that both the differentiation of monocytes into osteoclasts and the osteoclastic resorption of the calcium phosphate coating were significantly enhanced. This novel assay thus provides a new platform for studying osteoclastic activities and assessing drug efficacy in vitro.
Collapse
Affiliation(s)
- Sijia Chen
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Qiong Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Felipe Eltit
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| | - Yubin Guo
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Michael Cox
- Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H 3Z6, Canada
| | - Rizhi Wang
- Department of Materials Engineering, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
- School of Biomedical Engineering, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
- Centre for Hip Health and Mobility, University of British Columbia, Vancouver, BC V5Z 1M9, Canada
| |
Collapse
|
38
|
Delaisse JM, Søe K, Andersen TL, Rojek AM, Marcussen N. The Mechanism Switching the Osteoclast From Short to Long Duration Bone Resorption. Front Cell Dev Biol 2021; 9:644503. [PMID: 33859985 PMCID: PMC8042231 DOI: 10.3389/fcell.2021.644503] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 02/22/2021] [Indexed: 12/28/2022] Open
Abstract
The current models of osteoclastic bone resorption focus on immobile osteoclasts sitting on the bone surface and drilling a pit into the bone matrix. It recently appeared that many osteoclasts also enlarge their pit by moving across the bone surface while resorbing. Drilling a pit thus represents only the start of a resorption event of much larger amplitude. This prolonged resorption activity significantly contributes to pathological bone destruction, but the mechanism whereby the osteoclast engages in this process does not have an answer within the standard bone resorption models. Herein, we review observations that lead to envision how prolonged resorption is possible through simultaneous resorption and migration. According to the standard pit model, the “sealing zone” which surrounds the ruffled border (i.e., the actual resorption apparatus), “anchors” the ruffled border against the bone surface to be resorbed. Herein, we highlight that continuation of resorption demands that the sealing zone “glides” inside the cavity. Thereby, the sealing zone emerges as the structure responsible for orienting and displacing the ruffled border, e.g., directing resorption against the cavity wall. Importantly, sealing zone displacement stringently requires thorough collagen removal from the cavity wall - which renders strong cathepsin K collagenolysis indispensable for engagement of osteoclasts in cavity-enlargement. Furthermore, the sealing zone is associated with generation of new ruffled border at the leading edge, thereby allowing the ruffled border to move ahead. The sealing zone and ruffled border displacements are coordinated with the migration of the cell body, shown to be under control of lamellipodia at the leading edge and of the release of resorption products at the rear. We propose that bone resorption demands more attention to osteoclastic models integrating resorption and migration activities into just one cell phenotype.
Collapse
Affiliation(s)
- Jean-Marie Delaisse
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.,Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, Odense, Denmark.,Clinical Cell Biology, Pathology Research Unit, 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, Department of Pathology, Odense University Hospital, Odense, Denmark.,Clinical Cell Biology, Pathology Research Unit, 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
| | | | - Niels Marcussen
- Department of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
39
|
Jansen ID, Papapoulos SE, Bravenboer N, de Vries TJ, Appelman-Dijkstra NM. Increased Bone Resorption during Lactation in Pycnodysostosis. Int J Mol Sci 2021; 22:ijms22041810. [PMID: 33670411 PMCID: PMC7918824 DOI: 10.3390/ijms22041810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/04/2021] [Accepted: 02/07/2021] [Indexed: 12/03/2022] Open
Abstract
Pycnodysostosis, a rare autosomal recessive skeletal dysplasia, is caused by a deficiency of cathepsin K. Patients have impaired bone resorption in the presence of normal or increased numbers of multinucleated, but dysfunctional, osteoclasts. Cathepsin K degrades collagen type I and generates N-telopeptide (NTX) and the C-telopeptide (CTX) that can be quantified. Levels of these telopeptides are increased in lactating women and are associated with increased bone resorption. Nothing is known about the consequences of cathepsin K deficiency in lactating women. Here we present for the first time normalized blood and CTX measurements in a patient with pycnodysostosis, exclusively related to the lactation period. In vitro studies using osteoclasts derived from blood monocytes during lactation and after weaning further show consistent bone resorption before and after lactation. Increased expression of cathepsins L and S in osteoclasts derived from the lactating patient suggests that other proteinases could compensate for the lack of cathepsin K during the lactation period of pycnodysostosis patients.
Collapse
Affiliation(s)
- Ineke D.C. Jansen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands; (I.D.C.J.); (T.J.d.V.)
| | - Socrates E. Papapoulos
- Center for Bone Quality Department of Internal Medicine division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Nathalie Bravenboer
- Department of Clinical Chemistry, Amsterdam University Medical Center, Vrije Universiteit, 1081 HV Amsterdam, The Netherlands;
| | - Teun J. de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands; (I.D.C.J.); (T.J.d.V.)
| | - Natasha M. Appelman-Dijkstra
- Center for Bone Quality Department of Internal Medicine division of Endocrinology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
- Correspondence:
| |
Collapse
|
40
|
Abstract
PURPOSE OF REVIEW One aim in bone tissue engineering is to develop human cell-based, 3D in vitro bone models to study bone physiology and pathology. Due to the heterogeneity of cells among patients, patient's own cells are needed to be obtained, ideally, from one single cell source. This review attempts to identify the appropriate cell sources for development of such models. RECENT FINDINGS Bone marrow and peripheral blood are considered as suitable sources for extraction of osteoblast/osteocyte and osteoclast progenitor cells. Recent studies on these cell sources have shown no significant differences between isolated progenitor cells. However, various parameters such as medium composition affect the cell's proliferation and differentiation potential which could make the peripheral blood-derived stem cells superior to the ones from bone marrow. Peripheral blood can be considered a suitable source for osteoblast/osteocyte and osteoclast progenitor cells, being less invasive for the patient. However, more investigations are needed focusing on extraction and differentiation of both cell types from the same donor sample of peripheral blood.
Collapse
Affiliation(s)
- Sana Ansari
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands
| | - Sandra Hofmann
- Orthopaedic Biomechanics, Department of Biomedical Engineering and Institute for Complex Molecular Systems, Eindhoven University of Technology, P.O. Box 513, 5600 MB, Eindhoven, the Netherlands.
| |
Collapse
|
41
|
Heinemann C, Adam J, Kruppke B, Hintze V, Wiesmann HP, Hanke T. How to Get Them off?-Assessment of Innovative Techniques for Generation and Detachment of Mature Osteoclasts for Biomaterial Resorption Studies. Int J Mol Sci 2021; 22:ijms22031329. [PMID: 33572748 PMCID: PMC7865995 DOI: 10.3390/ijms22031329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 01/22/2021] [Accepted: 01/24/2021] [Indexed: 12/14/2022] Open
Abstract
The fusion process of mononuclear monocytes into multinuclear osteoclasts in vitro is an essential process for the study of osteoclastic resorption of biomaterials. Thereby biomaterials offer many influencing factors such as sample shape, material composition, and surface topography, which can have a decisive influence on the fusion and thus the entire investigation. For the specific investigation of resorption, it can therefore be advantageous to skip the fusion on samples and use mature, predifferentiated osteoclasts directly. However, most conventional detachment methods (cell scraper, accutase), lead to a poor survival rate of osteoclasts or to a loss of function of the cells after their reseeding. In the present study different conventional and novel methods of detachment in combination with different culture surfaces were investigated to obtain optimal osteoclast differentiation, yield, and vitality rates without loss of function. The innovative method-using thermoresponsive surfaces for cultivation and detachment-was found to be best suited. This is in particular due to its ability to maintain osteoclast activity, as proven by TRAP 5b-, CTSK-activity and resorption pits on dentin discs and decellularized osteoblast-derived matrix plates. In conclusion, it is shown, that osteoclasts can be predifferentiated on cell culture dishes and transferred to a reference biomaterial under preservation of osteoclastic resorption activity, providing biomaterial researchers with a novel tool for material characterization.
Collapse
|
42
|
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: 69] [Impact Index Per Article: 17.3] [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
|
43
|
Jourdain ML, Sergheraert J, Braux J, Guillaume C, Gangloff SC, Hubert D, Velard F, Jacquot J. Osteoclastogenesis and sphingosine-1-phosphate secretion from human osteoclast precursor monocytes are modulated by the cystic fibrosis transmembrane conductance regulator. Biochim Biophys Acta Mol Basis Dis 2020; 1867:166010. [PMID: 33188942 DOI: 10.1016/j.bbadis.2020.166010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 10/09/2020] [Accepted: 10/30/2020] [Indexed: 02/07/2023]
Abstract
Osteopenia and increased fracture rates are well-recognized in patients with cystic fibrosis (CF) disease. In CF pathology, F508del is the most common CFTR mutation, with more than 85% of patients carrying it on at least one allele. The underlying molecular defect in CFTR caused by the F508del-CFTR mutation in osteoclastogenesis, i.e., on the generation and bone-resorption activity of osteoclasts (OCs) from peripheral blood-derived monocytes (PBMCs) remained unexplored. We therefore investigated whether the F508del mutation could affect the osteoclastogenic capacity of PBMCs collected from 15 adult patients bearing the F508del-CFTR mutation, to modulate their bone-resorptive abilities and the level of sphingosine-1-phosphate (S1P) produced by OCs, a key factor in the bone mineral density and formation. In the present study, a severe, defective differentiation of CF-F508del PBMCs to CF-F508del OCs without any significant difference in nuclei number per OC was found compared to non-CF healthy PBMCs from 13 subjects after 7-14-days culture periods. We observed a reduced number of formed non-CF healthy OCs in the presence of a selective inhibitor of CFTR chloride conductance (CFTR-Inh172). Our data regarding OCs resorptive capabilites revealed that a loss of CFTR chloride activity in OCs led to a marked reduction in their trench-resorption mode. A 7-fold increase of the S1P release by CF-F508del OCs was found compared to non-CF healthy OCs after a 21-days culture period. We hypothesize that defective maturation of F508del-OCs precursor monocytes associated with high S1P production in the bone environment might contribute to low bone mineral density observed in the CF population.
Collapse
Affiliation(s)
- Marie-Laure Jourdain
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France; CHU de Reims, Pôle de Médecine Bucco-Dentaire, 51100 Reims, France
| | - Johan Sergheraert
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France; CHU de Reims, Pôle de Médecine Bucco-Dentaire, 51100 Reims, France
| | - Julien Braux
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France; CHU de Reims, Pôle de Médecine Bucco-Dentaire, 51100 Reims, France
| | - Christine Guillaume
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France
| | - Sophie C Gangloff
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France
| | - Dominique Hubert
- Service de Pneumologie, Adult Cystic Fibrosis Centre, Hôpital Cochin, 75012 Paris, France
| | - Frédéric Velard
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France.
| | - Jacky Jacquot
- Université de Reims Champagne Ardenne, BIOS EA 4691 "Biomatériaux et Inflammation en site osseux", 51097 Reims, France.
| |
Collapse
|
44
|
Møller AMJ, Delaisse J, Olesen JB, Bechmann T, Madsen JS, Søe K. Zoledronic Acid Is Not Equally Potent on Osteoclasts Generated From Different Individuals. JBMR Plus 2020; 4:e10412. [PMID: 33210064 PMCID: PMC7657394 DOI: 10.1002/jbm4.10412] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 08/27/2020] [Accepted: 09/10/2020] [Indexed: 12/23/2022] Open
Abstract
Zoledronic acid is a bisphosphonate commonly used to treat bone diseases such as osteoporosis and cancer‐induced bone disease. Patients exhibit a variable sensitivity to zoledronic acid; the underlying explanation for this remains unclear. The objective of this study was to obtain more knowledge in this regard. We hypothesized that osteoclasts generated from different individuals would show a variable sensitivity to zoledronic acid in vitro. Osteoclasts were generated using monocytes from 46 healthy female blood donors (40 to 66 years). Matured osteoclasts were reseeded onto bone slices precoated with different concentrations of zoledronic acid. IC50 values were determined based on total eroded bone surface after 3 days of resorption. The IC50 for inhibition of osteoclastic bone resorption varied from 0.06 to 12.57μM zoledronic acid; thus, a more than 200‐fold difference in sensitivity to zoledronic acid among osteoclasts from different individuals was observed. Multiple linear regression analyses showed that the determined IC50 correlated with smoking status, and the average number of nuclei per osteoclast in vitro. Further analyses showed that: (i) increasing protein levels of mature cathepsin K in osteoclast cultures rendered the osteoclasts less sensitive to zoledronic acid; (ii) surprisingly, neither the gene nor the protein expression of farnesyl diphosphate synthase was found to correlate with the IC50; and (iii) trench‐forming osteoclasts were found to be more sensitive to zoledronic acid than pit‐forming osteoclasts within the same cell culture. Thus, we conclude that there indeed is a high degree of variation in the potency of zoledronic acid on osteoclasts when generated from different individuals. We propose that our findings can explain some of the varying clinical efficacy of zoledronic acid therapy observed in patients, and may therefore be of clinical importance, which should be investigated in a clinical trial combining in vitro and in vivo investigations. © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Anaïs M J Møller
- Clinical Cell BiologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
- Department of Regional Health ResearchUniversity of Southern DenmarkVejleDenmark
- Clinical Cell Biology, Department of PathologyOdense University HospitalOdenseDenmark
- Department of Clinical Biochemistry and ImmunologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
| | - Jean‐Marie Delaisse
- Clinical Cell BiologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
- Department of Regional Health ResearchUniversity of Southern DenmarkVejleDenmark
- Clinical Cell Biology, Department of PathologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
| | - Jacob B Olesen
- Clinical Cell BiologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
- Clinical Cell Biology, Department of PathologyOdense University HospitalOdenseDenmark
| | - Troels Bechmann
- Department of Regional Health ResearchUniversity of Southern DenmarkVejleDenmark
- Department of OncologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
| | - Jonna S Madsen
- Department of Regional Health ResearchUniversity of Southern DenmarkVejleDenmark
- Department of Clinical Biochemistry and ImmunologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
| | - Kent Søe
- Clinical Cell BiologyLillebaelt Hospital, University Hospital of Southern DenmarkVejleDenmark
- Department of Regional Health ResearchUniversity of Southern DenmarkVejleDenmark
- Clinical Cell Biology, Department of PathologyOdense University HospitalOdenseDenmark
- Department of Clinical ResearchUniversity of Southern DenmarkOdenseDenmark
- Department of Molecular MedicineUniversity of Southern DenmarkOdenseDenmark
- OPEN, Open Patient data Explorative NetworkUniversity of Southern DenmarkOdenseDenmark
| |
Collapse
|
45
|
Borggaard XG, Pirapaharan DC, Delaissé JM, Søe K. Osteoclasts' Ability to Generate Trenches Rather Than Pits Depends on High Levels of Active Cathepsin K and Efficient Clearance of Resorption Products. Int J Mol Sci 2020; 21:ijms21165924. [PMID: 32824687 PMCID: PMC7460581 DOI: 10.3390/ijms21165924] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022] Open
Abstract
Until recently, it was well-accepted that osteoclasts resorb bone according to the resorption cycle model. This model is based on the assumption that osteoclasts are immobile during bone erosion, allowing the actin ring to be firmly attached and thereby provide an effective seal encircling the resorptive compartment. However, through time-lapse, it was recently documented that osteoclasts making elongated resorption cavities and trenches move across the bone surface while efficiently resorbing bone. However, it was also shown that osteoclasts making rounded cavities and pits indeed resorb bone while they are immobile. Only little is known about what distinguishes these two different resorption modes. This is of both basic and clinical interest because these resorption modes are differently sensitive to drugs and are affected by the gender as well as age of the donor. In the present manuscript we show that: 1. levels of active cathepsin K determine the switch from pit to trench mode; 2. pit and trench mode depend on clathrin-mediated endocytosis; and 3. a mechanism integrating release of resorption products and membrane/integrin recycling is required for prolongation of trench mode. Our study therefore contributes to an improved understanding of the molecular and cellular determinants for the two osteoclastic bone resorption modes.
Collapse
Affiliation(s)
- Xenia G. Borggaard
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
- Correspondence: (X.G.B.); (K.S.); Tel.: +45-65413190 (K.S.)
| | - Dinisha C. Pirapaharan
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Jean-Marie Delaissé
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
| | - Kent Søe
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, 7100 Vejle, Denmark; (D.C.P.); (J.-M.D.)
- Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5000 Odense C, Denmark
- Clinical Cell Biology, Department of Molecular Medicine, University of Southern Denmark, 5000 Odense C, Denmark
- Correspondence: (X.G.B.); (K.S.); Tel.: +45-65413190 (K.S.)
| |
Collapse
|
46
|
Møller AMJ, Delaissé JM, Olesen JB, Madsen JS, Canto LM, Bechmann T, Rogatto SR, Søe K. Aging and menopause reprogram osteoclast precursors for aggressive bone resorption. Bone Res 2020; 8:27. [PMID: 32637185 PMCID: PMC7329827 DOI: 10.1038/s41413-020-0102-7] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/06/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Women gradually lose bone from the age of ~35 years, but around menopause, the rate of bone loss escalates due to increasing bone resorption and decreasing bone formation levels, rendering these individuals more prone to developing osteoporosis. The increased osteoclast activity has been linked to a reduced estrogen level and other hormonal changes. However, it is unclear whether intrinsic changes in osteoclast precursors around menopause can also explain the increased osteoclast activity. Therefore, we set up a protocol in which CD14+ blood monocytes were isolated from 49 female donors (40-66 years old). Cells were differentiated into osteoclasts, and data on differentiation and resorption activity were collected. Using multiple linear regression analyses combining in vitro and in vivo data, we found the following: (1) age and menopausal status correlate with aggressive osteoclastic bone resorption in vitro; (2) the type I procollagen N-terminal propeptide level in vivo inversely correlates with osteoclast resorption activity in vitro; (3) the protein level of mature cathepsin K in osteoclasts in vitro increases with age and menopause; and (4) the promoter of the gene encoding the dendritic cell-specific transmembrane protein is less methylated with age. We conclude that monocytes are "reprogrammed" in vivo, allowing them to "remember" age, the menopausal status, and the bone formation status in vitro, resulting in more aggressive osteoclasts. Our discovery suggests that this may be mediated through DNA methylation. We suggest that this may have clinical implications and could contribute to understanding individual differences in age- and menopause-induced bone loss.
Collapse
Affiliation(s)
- Anaïs Marie Julie Møller
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Jean-Marie Delaissé
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
| | - Jacob Bastholm Olesen
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
| | - Jonna Skov Madsen
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Biochemistry and Immunology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Luisa Matos Canto
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Troels Bechmann
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Oncology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Silvia Regina Rogatto
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Clinical Genetics, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
| | - Kent Søe
- Clinical Cell Biology, Lillebaelt Hospital, University Hospital of Southern Denmark, 7100 Vejle, Denmark
- Department of Regional Health Research, University of Southern Denmark, 5230 Odense M, Denmark
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark
- Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark
- OPEN, Odense Patient data Explorative Network, Odense University Hospital, 5000 Odense C, Denmark
| |
Collapse
|
47
|
Søe K, Delaisse JM, Borggaard XG. Osteoclast formation at the bone marrow/bone surface interface: Importance of structural elements, matrix, and intercellular communication. Semin Cell Dev Biol 2020; 112:8-15. [PMID: 32563679 DOI: 10.1016/j.semcdb.2020.05.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/17/2020] [Accepted: 05/19/2020] [Indexed: 12/28/2022]
Abstract
Osteoclasts, the multinucleated cells responsible for bone resorption, have an enormous destructive power which demands to be kept under tight control. Accordingly, the identification of molecular signals directing osteoclastogenesis and switching on their resorptive activity have received much attention. Mandatory factors were identified, but a very essential aspect of the control mechanism of osteoclastic resorption, i.e. its spatial control, remains poorly understood. Under physiological conditions, multinucleated osteoclasts are only detected on the bone surface, while their mono-nucleated precursors are only in the bone marrow. How are pre-osteoclasts targeted to the bone surface? How is their progressive differentiation coordinated with their approach to the bone surface sites to be resorbed, which is where they finally fuse? Here we review the information on the bone marrow distribution of differentiating pre-osteoclasts relative to the position of the mandatory factors for their differentiation as well as relative to physical entities that may affect their access to the remodelling sites. This info allows recognizing an "osteoclastogenesis route" through the bone marrow and leading to the coincident fusion/resorption site - but also points to what still remains to be clarified regarding this route and regarding the restriction of fusion at the resorption site. Finally, we discuss the mechanism responsible for the start of resorption and its spatial extension. This review underscores that fully understanding the control of bone resorption requires to consider it in both space and time - which demands taking into account the context of bone tissue.
Collapse
Affiliation(s)
- Kent Søe
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark.
| | - Jean-Marie Delaisse
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark.
| | - Xenia Goldberg Borggaard
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense C, Denmark; Department of Clinical Research, University of Southern Denmark, 5230 Odense M, Denmark; Department of Molecular Medicine, University of Southern Denmark, 5230 Odense M, Denmark.
| |
Collapse
|
48
|
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: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [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
|
49
|
The matrix vesicle cargo miR-125b accumulates in the bone matrix, inhibiting bone resorption in mice. Commun Biol 2020; 3:30. [PMID: 31949279 PMCID: PMC6965124 DOI: 10.1038/s42003-020-0754-2] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 12/17/2019] [Indexed: 12/13/2022] Open
Abstract
Communication between osteoblasts and osteoclasts plays a key role in bone metabolism. We describe here an unexpected role for matrix vesicles (MVs), which bud from bone-forming osteoblasts and have a well-established role in initiation of bone mineralization, in osteoclastogenesis. We show that the MV cargo miR-125b accumulates in the bone matrix, with increased accumulation in transgenic (Tg) mice overexpressing miR-125b in osteoblasts. Bone formation and osteoblasts in Tg mice are normal, but the number of bone-resorbing osteoclasts is reduced, leading to higher trabecular bone mass. miR-125b in the bone matrix targets and degrades Prdm1, a transcriptional repressor of anti-osteoclastogenic factors, in osteoclast precursors. Overexpressing miR-125b in osteoblasts abrogates bone loss in different mouse models. Our results show that the MV cargo miR-125b is a regulatory element of osteoblast-osteoclast communication, and that bone matrix provides extracellular storage of miR-125b that is functionally active in bone resorption.
Collapse
|
50
|
Goel PN, Moharrer Y, Hebb JH, Egol AJ, Kaur G, Hankenson KD, Ahn J, Ashley JW. Suppression of Notch Signaling in Osteoclasts Improves Bone Regeneration and Healing. J Orthop Res 2019; 37:2089-2103. [PMID: 31166033 PMCID: PMC6739141 DOI: 10.1002/jor.24384] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 05/28/2019] [Indexed: 02/04/2023]
Abstract
Owing to the central role of osteoclasts in bone physiology and remodeling, manipulation of their maturation process provides a potential therapeutic strategy for treating bone diseases. To investigate this, we genetically inhibited the Notch signaling pathway in the myeloid lineage, which includes osteoclast precursors, using a dominant negative form of MAML (dnMAML) that inhibits the transcriptional complex required for downstream Notch signaling. Osteoclasts derived from dnMAML mice showed no significant differences in early osteoclastic gene expression compared to the wild type. Further, these demonstrated significantly lowered resorption activity using bone surfaces while retaining their osteoblast stimulating ability using ex vivo techniques. Using in vivo approaches, we detected significantly higher bone formation rates and osteoblast gene expression in dnMAML cohorts. Further, these mice exhibited increased bone/tissue mineral density compared to wild type and larger bony calluses in later stages of fracture healing. These observations suggest that therapeutic suppression of osteoclast Notch signaling could reduce, but not eliminate, osteoclastic resorption without suppression of restorative bone remodeling and, therefore, presents a balanced paradigm for increasing bone formation, regeneration, and healing. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 37:2089-2103, 2019.
Collapse
Affiliation(s)
- Peeyush N Goel
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Yasaman Moharrer
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - John H Hebb
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA,Georgetown University School of Medicine, Washington D.C
| | - Alexander J Egol
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | | | | | - Jaimo Ahn
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA,Corporal Michael J. Crescenz VA Medical Center, Philadelphia, PA,Co-corresponding Author: Jaimo Ahn, MD, PhD, FACS, FAOA, Advisory Dean, MSTP Steering Committee, Perelman School of Medicine, Co-Director, Orthopaedic Trauma, University of Pennsylvania Health System, Perelman School of Medicine, University of Pennsylvania, Investigator, Translational Musculoskeletal Research Center, Philadelphia Veterans Affairs Medical Center, 3737 Market Street, Floor 6, Philadelphia, PA-19104, Phone # +1 (215)-662-3340, Fax # +1 (215)-349-5890,
| | - Jason W Ashley
- Eastern Washington University, Cheney, WA,Corresponding Author: Jason Waid Ashley, PhD, Assistant Professor, Biology Department, 526 5th Street, SCI236, Eastern Washington University, Cheney, WA 99004, Phone # +1(509)-359-4665,
| |
Collapse
|