1
|
Maji S, Aliabouzar M, Quesada C, Chiravuri A, Macpherson A, Pinch A, Kazyak K, Emara Z, Abeid BA, Kent RN, Midekssa FS, Zhang M, Baker BM, Franceschi RT, Fabiilli ML. Ultrasound-generated bubbles enhance osteogenic differentiation of mesenchymal stromal cells in composite collagen hydrogels. Bioact Mater 2025; 43:82-97. [PMID: 39345992 PMCID: PMC11439547 DOI: 10.1016/j.bioactmat.2024.09.018] [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] [Received: 04/23/2024] [Revised: 08/30/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Hydrogels can improve the delivery of mesenchymal stromal cells (MSCs) by providing crucial biophysical cues that mimic the extracellular matrix. The differentiation of MSCs is dependent on biophysical cues like stiffness and viscoelasticity, yet conventional hydrogels cannot be dynamically altered after fabrication and implantation to actively direct differentiation. We developed a composite hydrogel, consisting of type I collagen and phase-shift emulsion, where osteogenic differentiation of MSCs can be non-invasively modulated using ultrasound. When exposed to ultrasound, the emulsion within the hydrogel was non-thermally vaporized into bubbles, which locally compacted and stiffened the collagen matrix surrounding each bubble. Bubble growth and matrix compaction were correlated, with collagen regions proximal (i.e., ≤ ∼60 μm) to the bubble displaying a 2.5-fold increase in Young's modulus compared to distal regions (i.e., > ∼60 μm). The viability and proliferation of MSCs, which were encapsulated within the composite hydrogel, were not impacted by bubble formation. In vitro and in vivo studies revealed encapsulated MSCs exhibited significantly elevated levels of RUNX2 and osteocalcin, markers of osteogenic differentiation, in collagen regions proximal to the bubble compared to distal regions. Additionally, alkaline phosphatase activity and calcium deposition were enhanced adjacent to the bubble. An opposite trend was observed for CD90, a marker of MSC stemness. Following subcutaneous implantation, bubbles persisted in the hydrogels for two weeks, which led to localized collagen alignment and increases in nuclear asymmetry. These results are a significant step toward controlling the 3D differentiation of MSCs in a non-invasive and on-demand manner.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Mitra Aliabouzar
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Carole Quesada
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Anjali Chiravuri
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aidan Macpherson
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Abigail Pinch
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Karsyn Kazyak
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Ziyad Emara
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Bachir A Abeid
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Robert N Kent
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Firaol S Midekssa
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Man Zhang
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Brendon M Baker
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Renny T Franceschi
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Periodontics and Oral Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Mario L Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Applied Physics Program, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
2
|
Sims NA. Osteoclast-derived coupling factors: origins and state-of-play Louis V Avioli lecture, ASBMR 2023. J Bone Miner Res 2024; 39:1377-1385. [PMID: 38990205 PMCID: PMC11425696 DOI: 10.1093/jbmr/zjae110] [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: 03/25/2024] [Revised: 06/28/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Coupling, the mechanism that controls the sequence of events in bone remodeling, is a fundamental theory for understanding the way the skeleton changes throughout life. This review is an adapted version of the Louis V Avioli lecture, delivered at the Annual Scientific Meeting of the American Society of Bone and Mineral Research in 2023. It outlines the history of the coupling concept, details how coupling is thought to occur within trabecular and cortical bone, and describes its multiple contexts and the many mechanisms suggested to couple bone-forming osteoblasts to the prior action of osteoclasts on the same bone surface. These mechanisms include signals produced at each stage of the remodeling sequence (resorption, reversal, and formation), such as factors released by osteoclasts through their resorptive action and through protein synthesis, molecules deposited in the cement line during the reversal phase, and potential signals from osteocytes within the local bone environment. The review highlights two examples of coupling factors (Cardiotrophin 1 and EphrinB2:EphB4) to illustrate the limited data available, the need to integrate the many functions of these factors within the basic multicellular unit (BMU), and the multiple origins of these factors, including the other cell types present during the remodeling sequence (such as osteocytes, macrophages, endothelial cells, and T-cells).
Collapse
Affiliation(s)
- Natalie A Sims
- Bone Cell Biology and Diease Unit, St. Vincent’s Institute of Medical Research, Fitzroy, Victoria 3065, Australia
- Department of Medicine at St. Vincent’s Hospital Melbourne, The University of Melbourne, Fitzroy, Victoria 3065, Australia
- The Mary McKillop Institute for Health Research, Australian Catholic University, Fitzroy, Victoria 3065, Australia
| |
Collapse
|
3
|
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
|
4
|
Kang J, Shibasaki M, Terauchi M, Oshibe N, Hyodo K, Marukawa E. Comparative analysis of the in vivo kinetic properties of various bone substitutes filled into a peri-implant canine defect model. J Periodontal Implant Sci 2024; 54:96-107. [PMID: 37857516 PMCID: PMC11065534 DOI: 10.5051/jpis.2204660233] [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: 11/11/2022] [Revised: 03/21/2023] [Accepted: 04/30/2023] [Indexed: 10/21/2023] Open
Abstract
PURPOSE Deproteinized bovine bone or synthetic hydroxyapatite are 2 prevalent bone grafting materials used in the clinical treatment of peri-implant bone defects. However, the differences in bone formation among these materials remain unclear. This study evaluated osteogenesis kinetics in peri-implant defects using 2 types of deproteinized bovine bone (Bio-Oss® and Bio-Oss/Collagen®) and 2 types of synthetic hydroxyapatite (Apaceram-AX® and Refit®). We considered factors including newly generated bone volume; bone, osteoid, and material occupancy; and bone-to-implant contact. METHODS A beagle model with a mandibular defect was created by extracting the bilateral mandibular third and fourth premolars. Simultaneously, an implant was inserted into the defect, and the space between the implant and the surrounding bone walls was filled with Bio-Oss, Bio-Oss/Collagen, Apaceram-AX, Refit, or autologous bone. Micro-computed tomography and histological analyses were conducted at 3 and 6 months postoperatively (Refit and autologous bone were not included at the 6-month time point due to their rapid absorption). RESULTS All materials demonstrated excellent biocompatibility and osteoconductivity. At 3 months, Bio-Oss and Apaceram-AX exhibited significantly greater volumes of formation than the other materials, with Bio-Oss having a marginally higher amount. However, this outcome was reversed at 6 months, with no significant difference between the 2 materials at either time point. Apaceram-AX displayed notably slower bioresorption and the largest quantity of residual material at both time points. In contrast, Refit had significantly greater bioresorption, with complete resorption and rapid maturation involving cortical bone formation at the crest at 3 months, Refit demonstrated the highest mineralized tissue and osteoid occupancy after 3 months, albeit without statistical significance. CONCLUSIONS Overall, the materials demonstrated varying post-implantation behaviors in vivo. Thus, in a clinical setting, both the properties of these materials and the specific conditions of the defects needing reinforcement should be considered to identify the most suitable material.
Collapse
Affiliation(s)
- Jingyang Kang
- Department of Regenerative and Reconstructive Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaki Shibasaki
- Department of Regenerative and Reconstructive Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.
| | - Masahiko Terauchi
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Narumi Oshibe
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuya Hyodo
- Department of Maxillofacial Surgery, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eriko Marukawa
- Department of Regenerative and Reconstructive Dentistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
5
|
Jeanneau C, Catherine JH, Giraud T, Lan R, About I. The Added Value of a Collagenated Thermosensitive Bone Substitute as a Scaffold for Bone Regeneration. MATERIALS (BASEL, SWITZERLAND) 2024; 17:625. [PMID: 38591482 PMCID: PMC10856128 DOI: 10.3390/ma17030625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/18/2024] [Accepted: 01/23/2024] [Indexed: 04/10/2024]
Abstract
A pre-hydrated thermosensitive collagenated biomaterial which sets at body temperature and maintains the space of the missing alveolar bone volume, OsteoBiol GTO® (GTO), has been released as a bone substitute. This study was designed to check its angiogenic and osteogenic potentials compared to OsteoBiol Gen-Os® (Gen-Os) and Geistlich Bio-Oss® (Bio-Oss). Samples of materials were incubated in culture media to obtain the extracts. Collagen release was measured in the extracts, which were used to investigate human periodontal ligament (hPDL) cell proliferation (MTT), colonization (Scratch assays) and growth factor release (ELISA). The effects on endothelial cell proliferation (MTT) and organization (Matrigel® assays) were also studied. Finally, endothelial and mesenchymal Stem Cell (hMSC) recruitment (Boyden Chambers) were investigated, and hMSC Alkaline Phosphatase (ALP) activity was measured. A higher collagen concentration was found in GTO extract, which led to significantly higher hPDL cell proliferation/colonization. All materials increased VEGF/FGF-2 growth factor secretion, endothelial cell recruitment, proliferation, and organization, but the increase was highest with GTO. All materials increased hMSC recruitment and ALP activity. However, the increase was highest with collagenated GTO and Gen-Os, which enhanced C5a and BMP-2 secretion. Overall, GTO has higher angiogenic/osteogenic potentials than the collagenated Gen-Os and the anorganic Bio-Oss. It provides a suitable scaffold for endothelial and mesenchymal stem cell recruitment, which represent essential bone regeneration requirements.
Collapse
Affiliation(s)
| | - Jean-Hugues Catherine
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France (J.-H.C.)
- APHM, Hôpital Timone, Pôle Odontologie, Service de Chirurgie Orale, 13005 Marseille, France;
| | - Thomas Giraud
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France (J.-H.C.)
- APHM, Hôpital Timone, Pôle Odontologie, Service de Chirurgie Orale, 13005 Marseille, France;
| | - Romain Lan
- APHM, Hôpital Timone, Pôle Odontologie, Service de Chirurgie Orale, 13005 Marseille, France;
- Aix-Marseille University, CNRS, EFS, ADES, 13385 Marseille, France
| | - Imad About
- Aix-Marseille University, CNRS, ISM, 13009 Marseille, France (J.-H.C.)
| |
Collapse
|
6
|
Kimira Y, Sato T, Sakamoto M, Osawa Y, Mano H. Collagen-Derived Dipeptide Pro-Hyp Enhanced ATDC5 Chondrocyte Differentiation under Hypoxic Conditions. Molecules 2023; 28:4664. [PMID: 37375217 DOI: 10.3390/molecules28124664] [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: 03/12/2023] [Revised: 05/26/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Chondrocytes are surrounded by a lower oxygen environment than other well-vascularized tissues with higher oxygenation levels. Prolyl-hydroxyproline (Pro-Hyp), one of the final collagen-derived peptides, has been previously reported to be involved in the early stages of chondrocyte differentiation. However, whether Pro-Hyp can alter chondrocyte differentiation under physiological hypoxic conditions is still unclear. This study aimed to investigate whether Pro-Hyp affects the differentiation of ATDC5 chondrogenic cells under hypoxic conditions. The addition of Pro-Hyp resulted in an approximately 18-fold increase in the glycosaminoglycan staining area compared to the control group under hypoxic conditions. Moreover, Pro-Hyp treatment significantly upregulated the expression of SOX9, Col2a1, Aggrecan, and MMP13 in chondrocytes cultured under hypoxic conditions. These results demonstrate that Pro-Hyp strongly promotes the early differentiation of chondrocytes under physiological hypoxic conditions. Therefore, Pro-Hyp, a bioactive peptide produced during collagen metabolism, may function as a remodeling factor or extracellular matrix remodeling signal that regulates chondrocyte differentiation in hypoxic cartilage.
Collapse
Affiliation(s)
- Yoshifumi Kimira
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi 350-0295, Saitama, Japan
| | - Takahiro Sato
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi 350-0295, Saitama, Japan
| | - Mayu Sakamoto
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi 350-0295, Saitama, Japan
| | - Yoshihiro Osawa
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi 350-0295, Saitama, Japan
| | - Hiroshi Mano
- Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Sciences, Josai University, 1-1 Keyakidai, Sakado-shi 350-0295, Saitama, Japan
| |
Collapse
|
7
|
Weivoda MM, Bradley EW. Macrophages and Bone Remodeling. J Bone Miner Res 2023; 38:359-369. [PMID: 36651575 PMCID: PMC10023335 DOI: 10.1002/jbmr.4773] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 01/05/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
Bone remodeling in the adult skeleton facilitates the removal and replacement of damaged and old bone to maintain bone quality. Tight coordination of bone resorption and bone formation during remodeling crucially maintains skeletal mass. Increasing evidence suggests that many cell types beyond osteoclasts and osteoblasts support bone remodeling, including macrophages and other myeloid lineage cells. Herein, we discuss the origin and functions for macrophages in the bone microenvironment, tissue resident macrophages, osteomacs, as well as newly identified osteomorphs that result from osteoclast fission. We also touch on the role of macrophages during inflammatory bone resorption. © 2023 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)
| | - Elizabeth W. Bradley
- Department of Orthopedics and Stem Cell Institute, University of Minnesota, Minneapolis, MN
| |
Collapse
|
8
|
Aguilera-Correa JJ, Gisbert-Garzarán M, Mediero A, Fernández-Aceñero MJ, de-Pablo-Velasco D, Lozano D, Esteban J, Vallet-Regí M. Antibiotic delivery from bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis caused by methicillin-resistant Staphylococcus aureus. Acta Biomater 2022; 154:608-625. [PMID: 36341887 DOI: 10.1016/j.actbio.2022.10.039] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 10/13/2022] [Accepted: 10/19/2022] [Indexed: 12/14/2022]
Abstract
Osteomyelitis is a hard-to-treat infection of the bone and bone marrow that is mainly caused by Staphylococcus aureus, with an increasing incidence of methicillin-resistant S. aureus (MRSA). Owing to the aggressiveness of these bacteria in colonizing and destroying the bone, systemic antibiotic treatments fail to eradicate the infection. Instead, it normally entails surgery to remove the dead or infected bone. In this work, we report bone-targeted mesoporous silica nanoparticles for the treatment of osteomyelitis. The nanoparticles have been engineered with a functional gelatine/colistin coating able to hamper premature release from the mesopores while effectively disaggregating the bacterial biofilm. Because antibiotic resistance is a global emergency, we have designed two sets of identical nanoparticles, carrying each of them a clinically relevant antibiotic, that have demonstrated to have synergistic effect. The bone-targeted nanoparticles have been thoroughly evaluated in vitro and in vivo, obtaining a notable reduction of the amount of bacteria in the bone in just 24 h after only one dose, and paving the way for localized, nanoparticle-mediated treatment of MRSA-caused osteomyelitis. STATEMENT OF SIGNIFICANCE: In this work, we propose the use of bone-targeted mesoporous silica nanoparticles to address S. aureus-caused osteomyelitis that render synergistic therapeutic effect via multidrug delivery. Because the bacterial biofilm is responsible for an aggressive surgical approach and prolonged antibiotic treatment, the nanoparticles have been functionalized with a functional coating able to both disaggregate the biofilm, hamper premature antibiotic release and protect the intact bone. These engineered nanoparticles are able to effectively target bone tissue both in vitro and in vivo, showing high biocompatibility and elevated antibacterial effect.
Collapse
Affiliation(s)
- J J Aguilera-Correa
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain
| | - M Gisbert-Garzarán
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - A Mediero
- Bone and Joint Unit, IIS- Fundación Jimenez Diaz, UAM, Avenida Reyes Católicos, 2 28037 Madrid, Spain
| | | | | | - D Lozano
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain
| | - J Esteban
- CIBER de Enfermedades Infecciosas (CIBERINFEC), 28029 Madrid, Spain; Clinical Microbiology Department, IIS-Fundación Jiménez Diaz, UAM, Avenida Reyes Católicos, 2 28037 Madrid, Spain.
| | - M Vallet-Regí
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre i+12, Plaza Ramón y Cajal s/n, 28040 Madrid, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), 28029 Madrid, Spain.
| |
Collapse
|
9
|
Chiu YL, Luo YL, Chen YW, Wu CT, Periasamy S, Yen KC, Hsieh DJ. Regenerative Efficacy of Supercritical Carbon Dioxide-Derived Bone Graft Putty in Rabbit Bone Defect Model. Biomedicines 2022; 10:2802. [PMID: 36359322 PMCID: PMC9687147 DOI: 10.3390/biomedicines10112802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/31/2022] [Accepted: 11/02/2022] [Indexed: 12/01/2023] Open
Abstract
Bone defects can arise from numerous reasons, such as aging, tumor, trauma, infection, surgery, and congenital diseases. Bone grafts are commonly used as a substitute to fill the void and regenerate the defect. Due to its clean and green technology, the supercritical carbon dioxide (SCCO2) extraction aided the production of bone grafts is a recent trend. The SCCO2-derived bone graft has osteoconductive and osteoinductive properties along with excellent biocompatible, nontoxic, bioabsorbable, osteoconductive, and good mechanical properties; however, clinical usage during surgery is time-consuming. Therefore, we produced a putty material combining bone graft powder and acellular dermal matrix (ADM) powder and tested its regenerative efficacy in the critical defect in the rabbit model. The putty was found to retain the tubular structure. In addition, the putty depicted excellent stickiness and cohesiveness in both saline and blood medium. The bone regeneration of bone graft and putty was similar; both had excellent bone healing and regeneration of critical defects as evaluated by the X-ray, microtomography, hematoxylin-eosin, Masson trichrome, and alizarin red staining. Putty contains a less washout rate, good mechanical strength, and biocompatibility. In conclusion, the SCCO2-derived moldable putty could be a promising easy-to-use alternative for bone grafts at present which might have real-world usage in orthopedics as a potential bone void filler and dental socket preservation.
Collapse
Affiliation(s)
- Yen-Lung Chiu
- Department of Life Sciences, National Cheng Kung University, Tainan 70101, Taiwan
| | - Yun-Li Luo
- Division of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei City 110, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei City 11490, Taiwan
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung 82151, Taiwan
| | - Yuan-Wu Chen
- Division of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei City 110, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei City 11490, Taiwan
| | - Chi-Tsung Wu
- Division of Oral and Maxillofacial Surgery, Tri-Service General Hospital, Taipei City 110, Taiwan
- School of Dentistry, National Defense Medical Center, Taipei City 11490, Taiwan
| | | | - Ko-Chung Yen
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung 82151, Taiwan
| | - Dar-Jen Hsieh
- R&D Center, ACRO Biomedical Co., Ltd., Kaohsiung 82151, Taiwan
| |
Collapse
|
10
|
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
|
11
|
Fu Z, Huang X, Zhou P, Wu B, Cheng L, Wang X, Zhu D. Protective effects of low-magnitude high-frequency vibration on high glucose-induced osteoblast dysfunction and bone loss in diabetic rats. J Orthop Surg Res 2021; 16:650. [PMID: 34717702 PMCID: PMC8557505 DOI: 10.1186/s13018-021-02803-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/18/2021] [Indexed: 01/07/2023] Open
Abstract
Objective Low-magnitude high-frequency vibration (LMHFV) has been reported to be capable of promoting osteoblast proliferation and differentiation. Reduced osteoblast activity and impaired bone formation were related to diabetic bone loss. We investigated the potential protective effects of LMHFV on high-glucose (HG)-induced osteoblasts in this study. In addition, the assessment of LMHFV treatment for bone loss attributed to diabetes was also performed in vivo.
Method MC3T3-E1 cells induced by HG only or treated with LMHFV were treated in vitro. The experiments performed in this study included the detection of cell proliferation, migration and differentiation, as well as protein expression. Diabetic bone loss induced by streptozotocin (STZ) in rats was established. Combined with bone morphometric, microstructure, biomechanical properties and matrix composition tests, the potential of LMHFV in treating diabetes bone loss was explored. Results After the application of LMHFV, the inhibiting effects of HG on the proliferation, migration and differentiation of osteoblasts were alleviated. The GSK3β/β-catenin pathway was involved in the protective effect of LMHFV. Impaired microstructure and biomechanical properties attributed to diabetes were ameliorated by LMHFV treatment. The improvement of femur biomechanical properties might be associated with the alteration of the matrix composition by the LMHFV. Conclusion LMHFV exhibited a protective effect on osteoblasts against HG by regulating the proliferation, migration and differentiation of osteoblasts. The function of promoting bone formation and reinforcing bone strength made it possible for LMHFV to alleviate diabetic bone loss. Supplementary Information The online version contains supplementary material available at 10.1186/s13018-021-02803-w.
Collapse
Affiliation(s)
- Zhaoyu Fu
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xu Huang
- Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Pengcheng Zhou
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Bo Wu
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Long Cheng
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinyu Wang
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Dong Zhu
- Department of Orthopaedic Trauma, The First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
12
|
Jerbić Radetić AT, Zoričić Cvek S, Tomas M, Erjavec I, Oguić M, Perić Kačarević Ž, Cvijanović Peloza O. CSBD Healing in Rats after Application of Bovine Xenogeneic Biomaterial Enriched with Magnesium Alloy. Int J Mol Sci 2021; 22:ijms22169089. [PMID: 34445794 PMCID: PMC8396602 DOI: 10.3390/ijms22169089] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Revised: 08/14/2021] [Accepted: 08/20/2021] [Indexed: 01/10/2023] Open
Abstract
Xenogeneic biomaterials Cerbone® and OsteoBiol® are widely used in oral implantology. In dental practice, xenogeneic biomaterial is usually combined with autologous bone to provide bone volume stability needed for long-term dental implants. Magnesium alloy implants dissolve and form mineral corrosion layer that is directly in contact with bone tissue, allowing deposition of the newly formed bone. CSBD heals by intramembranous ossification and therefore is a convenient model for analyses of ostoconductive and osteoinductive properties of different type of biomaterials. Magnesium alloy-enriched biomaterials have not yet been applied in oral implantology. Therefore, the aim of the current study was to investigate biological properties of potentially new bovine xenogeneic biomaterial enriched with magnesium alloy in a 5 mm CSBD model. Osteoconductive properties of Cerabone®, Cerabone® + Al. bone, and OsteoBiol® were also analyzed. Dynamics of bone healing was followed up on the days 3, 7, 15, 21, and 30. Calvary bone samples were analyzed by micro-CT, and values of the bone morphometric parameters were assessed. Bone samples were further processed for histological and immunohistochemical analyses. Histological observation revealed CSBD closure at day 30 of the given xenogeneic biomaterial groups, with the exception of the control group. TNF-α showed high intensity of expression at the sites of MSC clusters that underwent ossification. Osx was expressed in pre-osteoblasts, which were differentiated into mature osteoblasts and osteocytes. Results of the micro-CT analyses showed linear increase in bone volume of all xenogeneic biomaterial groups and also in the control. The highest average values of bone volume were found for the Cerabone® + Mg group. In addition, less residual biomaterial was estimated in the Cerabone® + Mg group than in the Cerabone® group, indicating its better biodegradation during CSBD healing. Overall, the magnesium alloy xenogeneic biomaterial demonstrated key properties of osteoinduction and biodegradidibility during CSBD healing, which is the reason why it should be recommended for application in clinical practice of oral implantology.
Collapse
Affiliation(s)
| | - Sanja Zoričić Cvek
- Department of Anatomy, Medical Faculty, University of Rijeka, 51 000 Rijeka, Croatia; (A.T.J.R.); (S.Z.C.)
| | - Matej Tomas
- Department of Dental Medicine, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, 31 000 Osijek, Croatia;
| | - Igor Erjavec
- Medical Faculty, University of Zagreb, 10 000 Zagreb, Croatia;
| | | | - Željka Perić Kačarević
- Department of Anatomy, Histology, Embryology, Pathology Anatomy and Pathology Histology, Faculty of Dental Medicine and Health Osijek, J.J. Strossmayer University of Osijek, 31 000 Osijek, Croatia;
| | - Olga Cvijanović Peloza
- Department of Anatomy, Medical Faculty, University of Rijeka, 51 000 Rijeka, Croatia; (A.T.J.R.); (S.Z.C.)
- Correspondence: ; Tel.: +385-91-8998-421
| |
Collapse
|
13
|
Nørregaard KS, Jürgensen HJ, Gårdsvoll H, Engelholm LH, Behrendt N, Søe K. Osteosarcoma and Metastasis Associated Bone Degradation-A Tale of Osteoclast and Malignant Cell Cooperativity. Int J Mol Sci 2021; 22:ijms22136865. [PMID: 34202300 PMCID: PMC8269025 DOI: 10.3390/ijms22136865] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 06/21/2021] [Accepted: 06/22/2021] [Indexed: 12/28/2022] Open
Abstract
Cancer-induced bone degradation is part of the pathological process associated with both primary bone cancers, such as osteosarcoma, and bone metastases originating from, e.g., breast, prostate, and colon carcinomas. Typically, this includes a cancer-dependent hijacking of processes also occurring during physiological bone remodeling, including osteoclast-mediated disruption of the inorganic bone component and collagenolysis. Extensive research has revealed the significance of osteoclast-mediated bone resorption throughout the course of disease for both primary and secondary bone cancer. Nevertheless, cancer cells representing both primary bone cancer and bone metastasis have also been implicated directly in bone degradation. We will present and discuss observations on the contribution of osteoclasts and cancer cells in cancer-associated bone degradation and reciprocal modulatory actions between these cells. The focus of this review is osteosarcoma, but we will also include relevant observations from studies of bone metastasis. Additionally, we propose a model for cancer-associated bone degradation that involves a collaboration between osteoclasts and cancer cells and in which both cell types may directly participate in the degradation process.
Collapse
Affiliation(s)
- Kirstine Sandal Nørregaard
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
- Correspondence: ; Tel.: +45-3545-6030
| | - Henrik Jessen Jürgensen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Henrik Gårdsvoll
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Lars Henning Engelholm
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center (BRIC), University of Copenhagen, 2200 Copenhagen, Denmark; (H.J.J.); (H.G.); (L.H.E.); (N.B.)
| | - Kent Søe
- Clinical Cell Biology, Pathology Research Unit, Department of Clinical Research, University of Southern Denmark, 5230 Odense, Denmark;
- Clinical Cell Biology, Department of Pathology, Odense University Hospital, 5000 Odense, Denmark
- Department of Molecular Medicine, University of Southern Denmark, 5230 Odense, Denmark
| |
Collapse
|
14
|
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
|
15
|
Collagenated Porcine Heterologous Bone Grafts: Histomorphometric Evaluation of Bone Formation Using Different Physical Forms in a Rabbit Cancellous Bone Model. Molecules 2021; 26:molecules26051339. [PMID: 33801547 PMCID: PMC7958959 DOI: 10.3390/molecules26051339] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/18/2021] [Accepted: 02/25/2021] [Indexed: 12/16/2022] Open
Abstract
Collagenated porcine-derived bone graft materials exhibit osteoconductive properties and the development of different formulations intends to enhance bone regeneration. This study aims to evaluate bone healing in a rabbit cancellous bone defect in response to grafting with different physicochemical forms of heterologous porcine bone. Twenty-six adult male New Zealand White rabbits received two critical size femoral bone defects per animal (n = 52), each randomly assigned to one of the five tested materials (Apatos, Gen-Os, mp3, Putty, and Gel 40). Animals were sacrificed at 15- and 30-days post-surgery. Qualitative and quantitative (new bone, particle and connective tissue percentages) histological analyses were performed. Histomorphometry showed statistically significant differences in all evaluated parameters between mp3 and both Putty and Gel 40 groups, regardless of the timepoint (p < 0.05). Moreover, statistical differences were observed between Apatos and both Putty (p = 0.014) and Gel 40 (p = 0.007) groups, at 30 days, in regard to particle percentage. Within each group, regarding new bone formation, mp3 showed significant differences (p = 0.028) between 15 (40.93 ± 3.49%) and 30 (52.49 ± 11.04%) days. Additionally, intragroup analysis concerning the percentage of particles revealed a significant reduction in particle occupied area from 15 to 30 days in mp3 and Gen-Os groups (p = 0.009). All mp3, Gen-Os and Apatos exhibited promising results in terms of new bone formation, thus presenting suitable alternatives to be used in bone regeneration.
Collapse
|
16
|
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
|
17
|
Guo P, Xue HY, Buttaro BA, Tran NT, Wong HL. Enhanced eradication of intracellular and biofilm-residing methicillin-resistant Staphylococcus aureus (MRSA) reservoirs with hybrid nanoparticles delivering rifampicin. Int J Pharm 2020; 589:119784. [PMID: 32877731 DOI: 10.1016/j.ijpharm.2020.119784] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 08/08/2020] [Accepted: 08/16/2020] [Indexed: 01/08/2023]
Abstract
Osteomyelitis carries a high risk of recurrence even after extended, aggressive antibiotic therapy. One of the key challenges is to eradicate the reservoirs of methicillin-resistant Staphylococcus aureus (MRSA) inside the host bone cells and their biofilms. Our goal is to develop rifampicin loaded lipid-polymer hybrid nanocarriers (Rf-LPN) and evaluate if they can achieve enhanced rifampicin delivery to eradicate these intracellular and biofilm-residing MRSA. After optimization of the composition, Rf-LPN demonstrated size around 110 nm in diameter that remained stable in serum-supplemented medium, drug payload up to 11.7% and sustained rifampicin release for 2 weeks. When comparing Rf-LPN with free rifampicin, moderate but significant (p < 0.05) improvement of the activities against three osteomyelitis-causing bacteria (USA300-0114, CDC-587, RP-62A) in planktonic form were observed. In comparison, the enhancements in the activities against the biofilms and intracellular MRSA by Rf-LPN were even more substantial. The MBEC50 values against USA300-0114, CDC-587, and RP-62A were 42 vs 155, 70 vs 388, and 265 ng/ml vs over 400 ng/ml, respectively, and up to 18.5-fold reduction in the intracellular MRSA counts in osteoblasts was obtained. Confocal microscope images confirmed extensive accumulation of Rf-LPN inside the biofilm matrix and MRSA-infected osteoblasts. Overall, in this proof-of-concept study we have developed and validated the strategy to exploit the nanoparticle-cell and nanoparticle-biofilm interactions with a new rifampicin nanoformulation for prevention of osteomyelitis recurrence and chronicity caused by the elusive MRSA.
Collapse
Affiliation(s)
- Pengbo Guo
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Hui Yi Xue
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Bettina A Buttaro
- Department of Microbiology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Ngoc T Tran
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA
| | - Ho Lun Wong
- School of Pharmacy, Temple University, 3307 North Broad Street, Philadelphia, PA 19140, USA.
| |
Collapse
|
18
|
Jürgensen HJ, van Putten S, Nørregaard KS, Bugge TH, Engelholm LH, Behrendt N, Madsen DH. Cellular uptake of collagens and implications for immune cell regulation in disease. Cell Mol Life Sci 2020; 77:3161-3176. [PMID: 32100084 PMCID: PMC11105017 DOI: 10.1007/s00018-020-03481-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 02/03/2020] [Accepted: 02/07/2020] [Indexed: 12/15/2022]
Abstract
As the dominant constituent of the extracellular matrix (ECM), collagens of different types are critical for the structural properties of tissues and make up scaffolds for cellular adhesion and migration. Importantly, collagens also directly modulate the phenotypic state of cells by transmitting signals that influence proliferation, differentiation, polarization, survival, and more, to cells of mesenchymal, epithelial, or endothelial origin. Recently, the potential of collagens to provide immune regulatory signals has also been demonstrated, and it is believed that pathological changes in the ECM shape immune cell phenotype. Collagens are themselves heavily regulated by a multitude of structural modulations or by catabolic pathways. One of these pathways involves a cellular uptake of collagens or soluble collagen-like defense collagens of the innate immune system mediated by endocytic collagen receptors. This cellular uptake is followed by the degradation of collagens in lysosomes. The potential of this pathway to regulate collagens in pathological conditions is evident from the increased extracellular accumulation of both collagens and collagen-like defense collagens following endocytic collagen receptor ablation. Here, we review how endocytic collagen receptors regulate collagen turnover during physiological conditions and in pathological conditions, such as fibrosis and cancer. Furthermore, we highlight the potential of collagens to regulate immune cells and discuss how endocytic collagen receptors can directly regulate immune cell activity in pathological conditions or do it indirectly by altering the extracellular milieu. Finally, we discuss the potential collagen receptors utilized by immune cells to directly detect ECM-related changes in the tissues which they encounter.
Collapse
Affiliation(s)
- Henrik J Jürgensen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark.
| | - Sander van Putten
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Kirstine S Nørregaard
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Lars H Engelholm
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Center, University of Copenhagen, Ole Maaloesvej 5, 2200, Copenhagen N, Denmark
| | - Daniel H Madsen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, 2730, Herlev, Denmark.
| |
Collapse
|
19
|
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
|
20
|
A Dynamic Hanging-Drop System for Mesenchymal Stem Cell Culture. Int J Mol Sci 2020; 21:ijms21124298. [PMID: 32560269 PMCID: PMC7352343 DOI: 10.3390/ijms21124298] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 06/02/2020] [Accepted: 06/13/2020] [Indexed: 02/06/2023] Open
Abstract
There have been many microfluid technologies combined with hanging-drop for cell culture gotten developed in the past decade. A common problem within these devices is that the cell suspension introduced at the central inlet could cause a number of cells in each microwell to not regularize. Also, the instability of droplets during the spheroid formation remains an unsolved ordeal. In this study, we designed a microfluidic-based hanging-drop culture system with the design of taper-tube that can increase the stability of droplets while enhancing the rate of liquid exchange. A ring is surrounding the taper-tube. The ring can hold the cells to enable us to seed an adequate amount of cells before perfusion. Moreover, during the period of cell culture, the mechanical force around the cell is relatively low to prevent stem cells from differentiate and maintain the phenotype. As a result of our hanging system design, cells are designed to accumulate at the bottom of the droplet. This method enhances convenience for observation activities and analysis of experiments. Thus, this microfluid chip can be used as an in vitro platform representing in vivo physiological conditions, and can be useful in regenerative therapy.
Collapse
|
21
|
Abourehab MAS. Hyaluronic Acid Modified Risedronate and Teriparatide Co-loaded Nanocarriers for Improved Osteogenic Differentiation of Osteoblasts for the Treatment of Osteoporosis. Curr Pharm Des 2020; 25:2975-2988. [PMID: 31368869 DOI: 10.2174/1381612825666190801140703] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022]
Abstract
BACKGROUND Owing to its multifactorial intricate pathogenesis, combined therapeutic regimen is considered appropriate for the treatment of osteoporosis. However, a multi-drug regimen is also associated with adverse effects due to the non-specific distribution of drugs. Therefore, the present study aims for efficient codelivery of risedronate (RDN) (a potent bone anti-resorptive drug) and teriparatide (TPD) (anabolic agent) as hyaluronic acid (HA)-modified chitosan nanoparticles (NPs). METHODS RDN/TPD NPs were synthesized using the high- pressure homogenization - solvent evaporation technique. The fabricated NPs were then characterized and optimized for suitable physicochemical characteristics. The optimized NPs were then evaluated for bone remodeling potential via assessment of time-mannered modulation in proliferation, differentiation, and mineralization of osteoblasts. RESULTS Results showed that HA-RDN/TPD NPs exhibited excellent physicochemical characteristics (nanoscopic size, stable zeta potential, high entrapment efficiency, and smooth spherical shape) and remained stable upon storage in the refrigerator. Assessment of various aspects of the cell growth cycle (i.e., proliferation, differentiation, and mineralization) evidenced promising bone regeneration efficacy of HA-RDN/TPD NPs. CONCLUSION This new strategy of employing simultaneous delivery of anti-resorptive and bone-forming agents would open new horizons for scientists, researchers, and healthcare providers as an efficient pharmacotherapy for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Mohammed A S Abourehab
- Department of Pharmaceutics, Faculty of Pharmacy, Minia University, Minia, Egypt.,Department of Pharmaceutics, Faculty of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| |
Collapse
|
22
|
Pahwa H, Khan MT, Sharan K. Hyperglycemia impairs osteoblast cell migration and chemotaxis due to a decrease in mitochondrial biogenesis. Mol Cell Biochem 2020; 469:109-118. [PMID: 32304005 DOI: 10.1007/s11010-020-03732-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 04/08/2020] [Indexed: 12/21/2022]
Abstract
Diabetes is associated with an increase in skeletal fragility and risk of fracture. However, the underlying mechanism for the same is not well understood. Specifically, the results from osteoblast cell culture studies are ambiguous due to contradicting reports. The use of supraphysiological concentrations in these studies, unachievable in vivo, might be the reason for the same. Therefore, here, we studied the effect of physiologically relevant levels of high glucose during diabetes (11.1 mM) on MC3T3-E1 osteoblast cell functions. The results showed that high glucose exposure to osteoblast cells increases their differentiation and mineralization without any effect on the proliferation. However, high glucose decreases their migratory potential and chemotaxis with a decrease in the associated cell signaling. Notably, this decrease in cell migration in high glucose conditions was accompanied by aberrant localization of Dynamin 2 in osteoblast cells. Besides, high glucose also caused a shift in mitochondrial dynamics towards the appearance of more fused and lesser fragmented mitochondria, with a concomitant decrease in the expression of DRP1, suggesting decreased mitochondrial biogenesis. In conclusion, here we are reporting for the first time that hyperglycemia causes a reduction in osteoblast cell migration and chemotaxis. This decrease might lead to an inefficient movement of osteoblasts to the erosion site resulting in uneven mineralization and skeletal fragility found in type 2 diabetes patients, in spite of having normal bone mineral density (BMD).
Collapse
Affiliation(s)
- Heena Pahwa
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
| | - Md Touseef Khan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India
| | - Kunal Sharan
- Department of Molecular Nutrition, CSIR-Central Food Technological Research Institute, Mysuru, 570020, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
23
|
Pirapaharan DC, Olesen JB, Andersen TL, Christensen SB, Kjærsgaard-Andersen P, Delaisse JM, Søe K. Catabolic activity of osteoblast lineage cells contributes to osteoclastic bone resorption in vitro. J Cell Sci 2019; 132:jcs.229351. [PMID: 30975918 DOI: 10.1242/jcs.229351] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 04/05/2019] [Indexed: 12/18/2022] Open
Abstract
Osteoblast lineage cells in human bone were recently shown to colonize eroded bone surfaces and to closely interact with osteoclasts. They proved to be identical to reversal cells and are believed to differentiate into bone-forming osteoblasts thereby coupling resorption and formation. However, they also exert catabolic activity that contributes to osteoclastic bone resorption, but this has not received much attention. Herein, we used co-cultures of primary human osteoblast lineage cells and human osteoclasts derived from peripheral blood monocytes to investigate whether a catabolic activity of osteoblast lineage cells could impact on osteoclastic bone resorption. Through a combination of immunofluorescence, in situ hybridization and time-lapse experiments, we show that MMP-13-expressing osteoblast lineage cells are attracted to and closely interact with bone-resorbing osteoclasts. This close interaction results in a strong and significant increase in the bone resorptive activity of osteoclasts - especially those making trenches. Importantly, we show that osteoclastic bone resorption becomes sensitive to inhibition of matrix metalloproteinases in the presence, but not in the absence, of osteoblast lineage cells. We propose that this may be due to the direct action of osteoblast-lineage-derived MMP-13 on bone resorption.
Collapse
Affiliation(s)
- Dinisha Cyril Pirapaharan
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Jacob Bastholm Olesen
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Sandra Bjerre Christensen
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | | | - Jean-Marie Delaisse
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| | - Kent Søe
- Department of Clinical Cell Biology, Vejle Hospital/Lillebaelt Hospital, Department of Regional Health Research, University of Southern Denmark, 7100 Vejle, Denmark
| |
Collapse
|
24
|
Dong Y, Yang L, Luo W, Zhu T, Yan W, Kong J, Yuan Z, Zhao Q. Mannose receptor C type 2 mediates 1,25(OH) 2D 3/vitamin D receptor-regulated collagen metabolism through collagen type 5, alpha 2 chain and matrix metalloproteinase 13 in murine MC3T3-E1 cells. Mol Cell Endocrinol 2019; 483:74-86. [PMID: 30641101 DOI: 10.1016/j.mce.2019.01.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 12/27/2018] [Accepted: 01/09/2019] [Indexed: 11/24/2022]
Abstract
Vitamin D plays an important role in maintaining skeletal development and bone homeostasis. Although vitamin D has been extensively researched, the direct effect of 1,25(OH)2D3 on osteoblasts is unclear. To explore the 1,25(OH)2D3 action on murine osteoblasts, we performed tandem mass tag experiments on MC3T3-E1 cells treated with and without 1,25(OH)2D3. Three up-regulated proteins (MRC2, WWTR1 and RASSF2) related to bone metabolism were confirmed in this study. 1,25(OH)2D3 up-regulated the expression of MRC2 through vitamin D receptor. MRC2 affects collagen metabolism in osteoblasts. Combined with bioinformatics and parallel reaction monitoring analysis, we inhibited the expression of MRC2 to explore the relationship between MRC2 and collagens. Then we found MRC2 down-regulated COL5A2 and up-regulated MMP13. This study provides a protein profile of 1,25(OH)2D3-treated murine osteoblasts, reveals a newly discovered signaling axis (1,25(OH)2D3/VDR/MRC2/COL5A2 and MMP13), and explains the effect of 1,25(OH)2D3 on bone metabolism from a new perspective.
Collapse
Affiliation(s)
- Yaping Dong
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Liping Yang
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Wenting Luo
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Tong Zhu
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Wei Yan
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China
| | - Juan Kong
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China; Nutrition Department, Shengjing Hospital of China Medical University, Shenyang, 110004, Liaoning, China
| | - Zhengwei Yuan
- Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China
| | - Qun Zhao
- Department of Pediatric Orthopedic, Shengjing Hospital of China Medical University, Shenyang, Liaoning, 110004, China; Key Laboratory of Health Ministry for Congenital Malformation, Shengjing Hospital of China Medical University, Benxi, Liaoning, 117004, China.
| |
Collapse
|
25
|
Sprangers S, Everts V. Molecular pathways of cell-mediated degradation of fibrillar collagen. Matrix Biol 2019; 75-76:190-200. [DOI: 10.1016/j.matbio.2017.11.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2017] [Revised: 10/06/2017] [Accepted: 11/09/2017] [Indexed: 12/12/2022]
|
26
|
Jürgensen HJ, Nørregaard KS, Sibree MM, Santoni-Rugiu E, Madsen DH, Wassilew K, Krustrup D, Garred P, Bugge TH, Engelholm LH, Behrendt N. Immune regulation by fibroblasts in tissue injury depends on uPARAP-mediated uptake of collectins. J Cell Biol 2018; 218:333-349. [PMID: 30366943 PMCID: PMC6314555 DOI: 10.1083/jcb.201802148] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Revised: 09/14/2018] [Accepted: 10/17/2018] [Indexed: 11/22/2022] Open
Abstract
Collectins such as mannose-binding lectin (MBL) and surfactant protein D (SP-D) become temporarily deposited in extravascular compartments after tissue injury and perform immune-stimulatory or inflammation-limiting functions. However, their turnover mechanisms, necessary to prevent excessive tissue damage, are virtually unknown. In this study, we show that fibroblasts in injured tissues undertake the clearance of collectins by using the endocytic collagen receptor uPARAP. In cellular assays, several types of collectins were endocytosed in a highly specific uPARAP-dependent process, not shared by the closely related receptor MR/CD206. When introduced into dermis or bleomycin-injured lungs of mice, collectins MBL and SP-D were endocytosed and routed for lysosomal degradation by uPARAP-positive fibroblasts. Fibroblast-specific expression of uPARAP governed endogenous SP-D levels and overall survival after lung injury. In lung tissue from idiopathic pulmonary fibrosis patients, a strong up-regulation of uPARAP was observed in fibroblasts adjacent to regions with SP-D secretion. This study demonstrates a novel immune-regulatory function of fibroblasts and identifies uPARAP as an endocytic receptor in immunity.
Collapse
Affiliation(s)
- Henrik J Jürgensen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.,Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Kirstine S Nørregaard
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Megan M Sibree
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Eric Santoni-Rugiu
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Daniel H Madsen
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark.,Center for Cancer Immune Therapy, Department of Haematology, Copenhagen University Hospital, Herlev, Denmark.,Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Katharina Wassilew
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Dorrit Krustrup
- Department of Pathology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Peter Garred
- Laboratory of Molecular Medicine, Department of Clinical Immunology Section 7631, Rigshospitalet, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thomas H Bugge
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD
| | - Lars H Engelholm
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| | - Niels Behrendt
- Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Nielsen CF, van Putten SM, Lund IK, Melander MC, Nørregaard KS, Jürgensen HJ, Reckzeh K, Christensen KR, Ingvarsen SZ, Gårdsvoll H, Jensen KE, Hamerlik P, Engelholm LH, Behrendt N. The collagen receptor uPARAP/Endo180 as a novel target for antibody-drug conjugate mediated treatment of mesenchymal and leukemic cancers. Oncotarget 2018; 8:44605-44624. [PMID: 28574834 PMCID: PMC5546505 DOI: 10.18632/oncotarget.17883] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2016] [Accepted: 04/24/2017] [Indexed: 11/29/2022] Open
Abstract
A key task in developing the field of personalized cancer therapy is the identification of novel molecular targets that enable treatment of cancers not susceptible to other means of specific therapy. The collagen receptor uPARAP/Endo180 is overexpressed by malignant cells in several non-epithelial cancers, notably including sarcomas, glioblastomas and subsets of acute myeloid leukemia. In contrast, in healthy adult individuals, expression is restricted to minor subsets of mesenchymal cells. Functionally, uPARAP/Endo180 is a rapidly recycling endocytic receptor that delivers its cargo directly into the endosomal-lysosomal system, thus opening a potential route of entry into receptor-positive cells. This combination of specific expression and endocytic function appears well suited for targeting of uPARAP/Endo180-positive cancers by antibody-drug conjugate (ADC) mediated drug delivery. Therefore, we utilized a specific monoclonal antibody against uPARAP/Endo180, raised through immunization of a uPARAP/Endo180 knock-out mouse, which reacts with both the human and the murine receptor, to construct a uPARAP-directed ADC. This antibody was coupled to the highly toxic dolastatin derivative, monomethyl auristatin E, via a cathepsin-labile valine-citrulline linker. With this ADC, we show strong and receptor-dependent cytotoxicity in vitro in uPARAP/Endo180-positive cancer cell lines of sarcoma, glioblastoma and leukemic origin. Furthermore, we demonstrate the potency of the ADC in vivo in a xenograft mouse model with human uPARAP/Endo180-positive leukemic cells, obtaining a complete cure of all tested mice following intravenous ADC treatment with no sign of adverse effects. Our study identifies uPARAP/Endo180 as a promising target for novel therapy against several highly malignant cancer types.
Collapse
Affiliation(s)
- Christoffer Fagernæs Nielsen
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Sander Maarten van Putten
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Ida Katrine Lund
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Maria Carlsén Melander
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kirstine Sandal Nørregaard
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Henrik Jessen Jürgensen
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark.,Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, NIDCR, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kristian Reckzeh
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Kristine Rothaus Christensen
- Experimental Animal Models Section, Department of Veterinary Disease Biology, University of Copenhagen, DK-1871 Frederiksberg C, Denmark
| | - Signe Ziir Ingvarsen
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Henrik Gårdsvoll
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | | | - Petra Hamerlik
- Danish Cancer Society Research Center, DK-2100 Copenhagen Ø, Denmark
| | - Lars Henning Engelholm
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Niels Behrendt
- The Finsen Laboratory, Rigshospitalet, Biotech Research and Innovation Center (BRIC), Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| |
Collapse
|
28
|
Patients' osteometabolic control improves the management of medication-related osteonecrosis of the jaw. Oral Surg Oral Med Oral Pathol Oral Radiol 2018; 125:147-156. [DOI: 10.1016/j.oooo.2017.10.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2017] [Revised: 09/03/2017] [Accepted: 10/18/2017] [Indexed: 12/18/2022]
|
29
|
Lassen NE, Andersen TL, Pløen GG, Søe K, Hauge EM, Harving S, Eschen GET, Delaisse JM. Coupling of Bone Resorption and Formation in Real Time: New Knowledge Gained From Human Haversian BMUs. J Bone Miner Res 2017; 32:1395-1405. [PMID: 28177141 DOI: 10.1002/jbmr.3091] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/20/2017] [Accepted: 01/30/2017] [Indexed: 01/31/2023]
Abstract
It is well known that bone remodeling starts with a resorption event and ends with bone formation. However, what happens in between and how resorption and formation are coupled remains mostly unknown. Remodeling is achieved by so-called basic multicellular units (BMUs), which are local teams of osteoclasts, osteoblasts, and reversal cells recently proven identical with osteoprogenitors. Their organization within a BMU cannot be appropriately analyzed in common histology. The originality of the present study is to capture the events ranging from initiation of resorption to onset of formation as a functional continuum. It was based on the position of specific cell markers in longitudinal sections of Haversian BMUs generating new canals through human long bones. It showed that initial resorption at the tip of the canal is followed by a period where newly recruited reversal/osteoprogenitor cells and osteoclasts alternate, thus revealing the existence of a mixed "reversal-resorption" phase. Three-dimensional reconstructions obtained from serial sections indicated that initial resorption is mainly involved in elongating the canal and the additional resorption events in widening it. Canal diameter measurements show that the latter contribute the most to overall resorption. Of note, the density of osteoprogenitors continuously grew along the "reversal/resorption" surface, reaching at least 39 cells/mm on initiation of bone formation. This value was independent of the length of the reversal/resorption surface. These observations strongly suggest that bone formation is initiated only above a threshold cell density, that the length of the reversal/resorption period depends on how fast osteoprogenitor recruitment reaches this threshold, and thus that the slower the rate of osteoprogenitor recruitment, the more bone is degraded. They lead to a model where the newly recognized reversal/resorption phase plays a central role in the mechanism linking osteoprogenitor recruitment and the resorption-formation switch. © 2017 American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Nicolai Ernlund Lassen
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | - Thomas Levin Andersen
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | - Gro Grunnet Pløen
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | - Kent Søe
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| | | | - Søren Harving
- Department of Orthopaedic Surgery, Aalborg University Hospital, Aalborg, Denmark
| | | | - Jean-Marie Delaisse
- Clinical Cell Biology, Institute of Regional Health Research, University of Southern Denmark, Vejle/Lillebaelt Hospital, Vejle, Denmark
| |
Collapse
|
30
|
Thiel A, Reumann MK, Boskey A, Wischmann J, von Eisenhart-Rothe R, Mayer-Kuckuk P. Osteoblast migration in vertebrate bone. Biol Rev Camb Philos Soc 2017. [PMID: 28631442 DOI: 10.1111/brv.12345] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Bone formation, for example during bone remodelling or fracture repair, requires mature osteoblasts to deposit bone with remarkable spatial precision. As osteoblast precursors derive either from circulation or resident stem cell pools, they and their progeny are required to migrate within the three-dimensional bone space and to navigate to their destination, i.e. to the site of bone formation. An understanding of this process is emerging based on in vitro and in vivo studies of several vertebrate species. Receptors on the osteoblast surface mediate cell adhesion and polarization, which induces osteoblast migration. Osteoblast migration is then facilitated along gradients of chemoattractants. The latter are secreted or released proteolytically by several cell types interacting with osteoblasts, including osteoclasts and vascular endothelial cells. The positions of these cellular sources of chemoattractants in relation to the position of the osteoblasts provide the migrating osteoblasts with tracks to their destination, and osteoblasts possess the means to follow a track marked by multiple chemoattractant gradients. In addition to chemotactic cues, osteoblasts sense other classes of signals and utilize them as landmarks for navigation. The composition of the osseous surface guides adhesion and hence migration efficiency and can also provide steering through haptotaxis. Further, it is likely that signals received from surface interactions modulate chemotaxis. Besides the nature of the surface, mechanical signals such as fluid flow may also serve as navigation signals for osteoblasts. Alterations in osteoblast migration and navigation might play a role in metabolic bone diseases such as osteoporosis.
Collapse
Affiliation(s)
- Antonia Thiel
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| | - Marie K Reumann
- Siegfried Weller Institute, BG Hospital, University of Tübingen, Schnarrenbergstraße 95, 72076 Tübingen, Germany
| | - Adele Boskey
- Mineralized Tissue Laboratory, Research Division, Hospital for Special Surgery, 535 E 70th Street, New York, NY 10021, U.S.A
| | - Johannes Wischmann
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| | - Rüdiger von Eisenhart-Rothe
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| | - Philipp Mayer-Kuckuk
- Bone Cell and Imaging Laboratory, Department of Orthopedics, Klinikum rechts der Isar, Ismaninger Straße 22, Technical University Munich, 81675 München, Germany
| |
Collapse
|
31
|
Internalization of Collagen: An Important Matrix Turnover Pathway in Cancer. EXTRACELLULAR MATRIX IN TUMOR BIOLOGY 2017. [DOI: 10.1007/978-3-319-60907-2_2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
32
|
Bone Formation from Porcine Dental Germ Stem Cells on Surface Modified Polybutylene Succinate Scaffolds. Stem Cells Int 2016; 2016:8792191. [PMID: 27413380 PMCID: PMC4927991 DOI: 10.1155/2016/8792191] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 05/05/2016] [Accepted: 05/16/2016] [Indexed: 01/09/2023] Open
Abstract
Designing and providing a scaffold are very important for the cells in tissue engineering. Polybutylene succinate (PBS) has high potential as a scaffold for bone regeneration due to its capacity in cell proliferation and differentiation. Also, stem cells from 3rd molar tooth germs were favoured in this study due to their developmentally and replicatively immature nature. In this study, porcine dental germ stem cells (pDGSCs) seeded PBS scaffolds were used to investigate the effects of surface modification with fibronectin or laminin on these scaffolds to improve cell attachment, proliferation, and osteogenic differentiation for tissue engineering applications. The osteogenic potentials of pDGSCs on these modified and unmodified foams were examined to heal bone defects and the effects of fibronectin or laminin modified PBS scaffolds on pDGSC differentiation into bone were compared for the first time. For this study, MTS assay was used to assess the cytotoxic effects of modified and unmodified surfaces. For the characterization of pDGSCs, flow cytometry analysis was carried out. Besides, alkaline phosphatase (ALP) assay, von Kossa staining, real-time PCR, CM-Dil, and immunostaining were applied to analyze osteogenic potentials of pDGSCs. The results of these studies demonstrated that pDGSCs were differentiated into osteogenic cells on fibronectin modified PBS foams better than those on unmodified and laminin modified PBS foams.
Collapse
|
33
|
Guided bone regeneration is promoted by the molecular events in the membrane compartment. Biomaterials 2016; 84:167-183. [DOI: 10.1016/j.biomaterials.2016.01.034] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 01/18/2016] [Indexed: 11/18/2022]
|
34
|
Abdelgawad ME, Delaisse JM, Hinge M, Jensen PR, Alnaimi RW, Rolighed L, Engelholm LH, Marcussen N, Andersen TL. Early reversal cells in adult human bone remodeling: osteoblastic nature, catabolic functions and interactions with osteoclasts. Histochem Cell Biol 2016; 145:603-15. [PMID: 26860863 DOI: 10.1007/s00418-016-1414-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/15/2015] [Indexed: 12/31/2022]
Abstract
The mechanism coupling bone resorption and formation is a burning question that remains incompletely answered through the current investigations on osteoclasts and osteoblasts. An attractive hypothesis is that the reversal cells are likely mediators of this coupling. Their nature is a big matter of debate. The present study performed on human cancellous bone is the first one combining in situ hybridization and immunohistochemistry to demonstrate their osteoblastic nature. It shows that the Runx2 and CD56 immunoreactive reversal cells appear to take up TRAcP released by neighboring osteoclasts. Earlier preclinical studies indicate that reversal cells degrade the organic matrix left behind by the osteoclasts and that this degradation is crucial for the initiation of the subsequent bone formation. To our knowledge, this study is the first addressing these catabolic activities in adult human bone through electron microscopy and analysis of molecular markers. Periosteoclastic reversal cells show direct contacts with the osteoclasts and with the demineralized resorption debris. These early reversal cells show (1) ¾-collagen fragments typically generated by extracellular collagenases of the MMP family, (2) MMP-13 (collagenase-3) and (3) the endocytic collagen receptor uPARAP/Endo180. The prevalence of these markers was lower in the later reversal cells, which are located near the osteoid surfaces and morphologically resemble mature bone-forming osteoblasts. In conclusion, this study demonstrates that reversal cells colonizing bone surfaces right after resorption are osteoblast-lineage cells, and extends to adult human bone remodeling their role in rendering eroded surfaces osteogenic.
Collapse
Affiliation(s)
- Mohamed Essameldin Abdelgawad
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.,Faculty of Science, Helwan University, Helwan, Egypt
| | - Jean-Marie Delaisse
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.
| | - Maja Hinge
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.,Division of Hematology, Department of Internal Medicine, Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Vejle, Denmark
| | - Pia Rosgaard Jensen
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark
| | - Ragad Walid Alnaimi
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark
| | - Lars Rolighed
- Breast and Endocrine Section, Department of Surgery, Aarhus University Hospital, Aarhus, Denmark
| | - Lars H Engelholm
- The Finsen Laboratory, Rigshospitalet/Biotech Research and Innovation Centre (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Niels Marcussen
- Department of Clinical Pathology, Odense University Hospital, Odense, Denmark
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology (KCB), Vejle Hospital - Lillebaelt Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100, Vejle, Denmark.
| |
Collapse
|
35
|
Sturge J. Endo180 at the cutting edge of bone cancer treatment and beyond. J Pathol 2016; 238:485-8. [PMID: 26576691 PMCID: PMC4819699 DOI: 10.1002/path.4673] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2015] [Accepted: 11/14/2015] [Indexed: 12/21/2022]
Abstract
Skeletal bone is an attractive site for secondary tumour growth and is also home to spontaneous primary cancer. Treatment of bone metastasis is focused on limiting the vicious cycle of bone destruction with bisphosphonates or inhibition of receptor activator of nuclear factor‐κB ligand (RANKL) with the fully human monoclonal antibody denosumab. The estimated 1 million deaths/year where bone metastasis is present, and the high healthcare costs required for its management, have ignited intensive research into the cellular and molecular pathology of osteolysis, involving interplay between tumour cells, bone‐forming osteoblasts and bone‐degrading osteoclasts. Compared to bone metastasis, knowledge about the pathology of primary bone cancers is limited. In recent work published in this journal, Engelholm et al provide a unique insight into how this poorly understood disease manifests and destroys bone. For the first time they have demonstrated that a mouse monoclonal antibody targeting the collagen receptor Endo180 (CD280, MRC2 uPARAP) can prevent osteolysis and bone destruction in a syngeneic model of advanced osteosarcoma. Their convincing findings make an important contribution towards Endo180‐based therapy being developed as an option for the treatment of bone cancer amongst other malignancies. © 2015 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Justin Sturge
- School of Biological, Biomedical and Environmental Sciences, University of Hull, UK
| |
Collapse
|
36
|
Engelholm LH, Melander MC, Hald A, Persson M, Madsen DH, Jürgensen HJ, Johansson K, Nielsen C, Nørregaard KS, Ingvarsen SZ, Kjaer A, Trovik CS, Laerum OD, Bugge TH, Eide J, Behrendt N. Targeting a novel bone degradation pathway in primary bone cancer by inactivation of the collagen receptor uPARAP/Endo180. J Pathol 2015; 238:120-33. [PMID: 26466547 DOI: 10.1002/path.4661] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 09/08/2015] [Accepted: 10/08/2015] [Indexed: 11/09/2022]
Abstract
In osteosarcoma, a primary mesenchymal bone cancer occurring predominantly in younger patients, invasive tumour growth leads to extensive bone destruction. This process is insufficiently understood, cannot be efficiently counteracted and calls for novel means of treatment. The endocytic collagen receptor, uPARAP/Endo180, is expressed on various mesenchymal cell types and is involved in bone matrix turnover during normal bone growth. Human osteosarcoma specimens showed strong expression of this receptor on tumour cells, along with the collagenolytic metalloprotease, MT1-MMP. In advanced tumours with ongoing bone degeneration, sarcoma cells positive for these proteins formed a contiguous layer aligned with the degradation zones. Remarkably, osteoclasts were scarce or absent from these regions and quantitative analysis revealed that this scarcity marked a strong contrast between osteosarcoma and bone metastases of carcinoma origin. This opened the possibility that sarcoma cells might directly mediate bone degeneration. To examine this question, we utilized a syngeneic, osteolytic bone tumour model with transplanted NCTC-2472 sarcoma cells in mice. When analysed in vitro, these cells were capable of degrading the protein component of surface-labelled bone slices in a process dependent on MMP activity and uPARAP/Endo180. Systemic treatment of the sarcoma-inoculated mice with a mouse monoclonal antibody that blocks murine uPARAP/Endo180 led to a strong reduction of bone destruction. Our findings identify sarcoma cell-resident uPARAP/Endo180 as a central player in the bone degeneration of advanced tumours, possibly following an osteoclast-mediated attack on bone in the early tumour stage. This points to uPARAP/Endo180 as a promising therapeutic target in osteosarcoma, with particular prospects for improved neoadjuvant therapy.
Collapse
Affiliation(s)
- Lars H Engelholm
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Maria C Melander
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Andreas Hald
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Morten Persson
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Denmark
| | - Daniel H Madsen
- Proteases and Tissue Remodelling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Henrik J Jürgensen
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Kristina Johansson
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Christoffer Nielsen
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Kirstine S Nørregaard
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Signe Z Ingvarsen
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology, Nuclear Medicine and PET and Cluster for Molecular Imaging, Rigshospitalet and University of Copenhagen, Denmark
| | - Clement S Trovik
- Department of Oncology/Orthopaedics, Haukeland University Hospital, Bergen, Norway
| | - Ole D Laerum
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark.,Department of Clinical Medicine, Gade Laboratory of Pathology, University of Bergen, Norway
| | - Thomas H Bugge
- Proteases and Tissue Remodelling Section, National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, USA
| | - Johan Eide
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Niels Behrendt
- Finsen Laboratory/Biotech Research and Innovation Centre (BRIC), Rigshospitalet and University of Copenhagen, Denmark
| |
Collapse
|
37
|
Melander MC, Jürgensen HJ, Madsen DH, Engelholm LH, Behrendt N. The collagen receptor uPARAP/Endo180 in tissue degradation and cancer (Review). Int J Oncol 2015; 47:1177-88. [PMID: 26316068 PMCID: PMC4583827 DOI: 10.3892/ijo.2015.3120] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2015] [Accepted: 07/20/2015] [Indexed: 01/08/2023] Open
Abstract
The collagen receptor uPARAP/Endo180, the product of the MRC2 gene, is a central component in the collagen turnover process governed by various mesenchymal cells. Through the endocytosis of collagen or large collagen fragments, this recycling receptor serves to direct basement membrane collagen as well as interstitial collagen to lysosomal degradation. This capacity, shared only with the mannose receptor from the same protein family, endows uPARAP/Endo180 with a critical role in development and homeostasis, as well as in pathological disruptions of the extracellular matrix structure. Important pathological functions of uPARAP/Endo180 have been identified in various cancers and in several fibrotic conditions. With a particular focus on matrix turnover in cancer, this review presents the necessary background for understanding the function of uPARAP/Endo180 at the molecular and cellular level, followed by an in-depth survey of the available knowledge of the expression and role of this receptor in various types of cancer and other degenerative diseases.
Collapse
Affiliation(s)
- Maria C Melander
- The Finsen Laboratory, Rigshospitalet/BRIC, The University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Henrik J Jürgensen
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, NIDCR, National Institutes of Health, Bethesda, MD, USA
| | - Daniel H Madsen
- Proteases and Tissue Remodeling Section, Oral and Pharyngeal Cancer Branch, NIDCR, National Institutes of Health, Bethesda, MD, USA
| | - Lars H Engelholm
- The Finsen Laboratory, Rigshospitalet/BRIC, The University of Copenhagen, DK-2200 Copenhagen N, Denmark
| | - Niels Behrendt
- The Finsen Laboratory, Rigshospitalet/BRIC, The University of Copenhagen, DK-2200 Copenhagen N, Denmark
| |
Collapse
|
38
|
Jensen PR, Andersen TL, Hauge EM, Bollerslev J, Delaissé JM. A joined role of canopy and reversal cells in bone remodeling--lessons from glucocorticoid-induced osteoporosis. Bone 2015; 73:16-23. [PMID: 25497571 DOI: 10.1016/j.bone.2014.12.004] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Revised: 09/19/2014] [Accepted: 12/04/2014] [Indexed: 12/18/2022]
Abstract
Successful bone remodeling demands that osteoblasts restitute the bone removed by osteoclasts. In human cancellous bone, a pivotal role in this restitution is played by the canopies covering the bone remodeling surfaces, since disruption of canopies in multiple myeloma, postmenopausal- and glucocorticoid-induced osteoporosis is associated with the absence of progression of the remodeling cycle to bone formation, i.e., uncoupling. An emerging concept explaining this critical role of canopies is that they represent a reservoir of osteoprogenitors to be delivered to reversal surfaces. In postmenopausal osteoporosis, this concept is supported by the coincidence between the absence of canopies and scarcity of cells on reversal surfaces together with abortion of the remodeling cycle. Here we tested whether this concept holds true in glucocorticoid-induced osteoporosis. A histomorphometric analysis of iliac crest biopsies from patients exposed to long-term glucocorticoid treatment revealed a subpopulation of reversal surfaces corresponding to the characteristics of arrest found in postmenopausal osteoporosis. Importantly, these arrested reversal surfaces were devoid of canopy coverage in almost all biopsies, and their prevalence correlated with a deficiency in bone forming surfaces. Taken together with the other recent data, the functional link between canopies, reversal surface activity, and the extent of bone formation surface in postmenopausal- and glucocorticoid-induced osteoporosis, supports a model where bone restitution during remodeling demands recruitment of osteoprogenitors from the canopy onto reversal surfaces. These data suggest that securing the presence of functional local osteoprogenitors deserves attention in the search of strategies to prevent the bone loss that occurs during bone remodeling in pathological situations.
Collapse
Affiliation(s)
- Pia Rosgaard Jensen
- Department of Clinical Cell Biology (KCB), Vejle Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100 Vejle, Denmark.
| | - Thomas Levin Andersen
- Department of Clinical Cell Biology (KCB), Vejle Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100 Vejle, Denmark
| | - Ellen-Margrethe Hauge
- Department of Rheumatology, Aarhus University Hospital, Building 3, Nørrebrogade 44, 8000 Aarhus C, Denmark
| | - Jens Bollerslev
- Section of Specialized Endocrinology, Medical Clinic B, Oslo University Hospital, Oslo, Norway; Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Jean-Marie Delaissé
- Department of Clinical Cell Biology (KCB), Vejle Hospital, IRS, University of Southern Denmark, Kabbeltoft 25, 7100 Vejle, Denmark
| |
Collapse
|