1
|
Doms S, Verlinden L, Janssens I, Vanhevel J, Eerlings R, Houtman R, Kato S, Mathieu C, Decallonne B, Carmeliet G, Verstuyf A. Coactivator-independent vitamin D receptor signaling causes severe rickets in mice, that is not prevented by a diet high in calcium, phosphate, and lactose. Bone Res 2024; 12:44. [PMID: 39164247 PMCID: PMC11335873 DOI: 10.1038/s41413-024-00343-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/26/2024] [Accepted: 05/12/2024] [Indexed: 08/22/2024] Open
Abstract
The vitamin D receptor (VDR) plays a critical role in the regulation of mineral and bone homeostasis. Upon binding of 1α,25-dihydroxyvitamin D3 to the VDR, the activation function 2 (AF2) domain repositions and recruits coactivators for the assembly of the transcriptional machinery required for gene transcription. In contrast to coactivator-induced transcriptional activation, the functional effects of coactivator-independent VDR signaling remain unclear. In humans, mutations in the AF2 domain are associated with hereditary vitamin D-resistant rickets, a genetic disorder characterized by impaired bone mineralization and growth. In the present study, we used mice with a systemic or conditional deletion of the VDR-AF2 domain (VdrΔAF2) to study coactivator-independent VDR signaling. We confirm that ligand-induced transcriptional activation was disabled because the mutant VDRΔAF2 protein was unable to interact with coactivators. Systemic VdrΔAF2 mice developed short, undermineralized bones with dysmorphic growth plates, a bone phenotype that was more pronounced than that of systemic Vdr knockout (Vdr-/-) mice. Interestingly, a rescue diet that is high in calcium, phosphate, and lactose, normalized this phenotype in Vdr-/-, but not in VdrΔAF2 mice. However, osteoblast- and osteoclast-specific VdrΔAF2 mice did not recapitulate this bone phenotype indicating coactivator-independent VDR effects are more important in other organs. In addition, RNA-sequencing analysis of duodenum and kidney revealed a decreased expression of VDR target genes in systemic VdrΔAF2 mice, which was not observed in Vdr-/- mice. These genes could provide new insights in the compensatory (re)absorption of minerals that are crucial for bone homeostasis. In summary, coactivator-independent VDR effects contribute to mineral and bone homeostasis.
Collapse
Affiliation(s)
- Stefanie Doms
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Lieve Verlinden
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Iris Janssens
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Justine Vanhevel
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Roy Eerlings
- Department of Cellular and Molecular Medicine, Laboratory of Molecular Endocrinology, KU Leuven, Leuven, Belgium
- Institute of Applied Microbiology, RWTH Aachen University, Aachen, Germany
| | | | - Shigeaki Kato
- Health Sciences Research Center, Iryo Sosei University, Iwaki, Fukuchima, Japan
- Research Institute of Innovative Medicine, Tokiwa Foundation, Iwaki, Fukuchima, Japan
| | - Chantal Mathieu
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Brigitte Decallonne
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Geert Carmeliet
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium
| | - Annemieke Verstuyf
- Department of Chronic diseases and metabolism, Laboratory of Clinical and Experimental Endocrinology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
2
|
Ceylan M, Schoenmaker T, Hogervorst JMA, Jansen IDC, Schimmel IM, Prins CM, Laine ML, de Vries TJ. Osteogenic Differentiation of Human Gingival Fibroblasts Inhibits Osteoclast Formation. Cells 2024; 13:1090. [PMID: 38994943 PMCID: PMC11240541 DOI: 10.3390/cells13131090] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/13/2024] Open
Abstract
Gingival fibroblasts (GFs) can differentiate into osteoblast-like cells and induce osteoclast precursors to differentiate into osteoclasts. As it is unclear whether these two processes influence each other, we investigated how osteogenic differentiation of GFs affects their osteoclast-inducing capacity. To establish step-wise mineralization, GFs were cultured in four groups for 3 weeks, without or with osteogenic medium for the final 1, 2, or all 3 weeks. The mineralization was assessed by ALP activity, calcium concentration, scanning electron microscopy (SEM), Alizarin Red staining, and quantitative PCR (qPCR). To induce osteoclast differentiation, these cultures were then co-cultured for a further 3 weeks with peripheral blood mononuclear cells (PBMCs) containing osteoclast precursors. Osteoclast formation was assessed at different timepoints with qPCR, enzyme-linked immunosorbent assay (ELISA), TRAcP activity, and staining. ALP activity and calcium concentration increased significantly over time. As confirmed with the Alizarin Red staining, SEM images showed that the mineralization process occurred over time. Osteoclast numbers decreased in the GF cultures that had undergone osteogenesis. TNF-α secretion, a costimulatory molecule for osteoclast differentiation, was highest in the control group. GFs can differentiate into osteoblast-like cells and their degree of differentiation reduces their osteoclast-inducing capacity, indicating that, with appropriate stimulation, GFs could be used in regenerative periodontal treatments.
Collapse
Affiliation(s)
- Merve Ceylan
- Department of Periodontology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Ton Schoenmaker
- Department of Periodontology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Jolanda M A Hogervorst
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Ineke D C Jansen
- Department of Periodontology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Irene M Schimmel
- Department of Medical Biology, Amsterdam University Medical Centers, Location AMC, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Caya M Prins
- Department of Periodontology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Marja L Laine
- Department of Periodontology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| | - Teun J de Vries
- Department of Periodontology, Academic Centre for Dentistry Amsterdam, University of Amsterdam and Vrije University Amsterdam, Gustav Mahlerlaan 3004, 1081 LA Amsterdam, The Netherlands
| |
Collapse
|
3
|
Kanno Y. The Roles of Fibrinolytic Factors in Bone Destruction Caused by Inflammation. Cells 2024; 13:516. [PMID: 38534360 PMCID: PMC10968824 DOI: 10.3390/cells13060516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 03/14/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis, spondyloarthritis, systemic lupus erythematosus, Crohn's disease, periodontitis, and carcinoma metastasis frequently result in bone destruction. Pro-inflammatory cytokines such as tumor necrosis factor-α (TNF-α), interleukin-1β (IL-1β), IL-6, and IL-17 are known to influence bone loss by promoting the differentiation and activation of osteoclasts. Fibrinolytic factors, such as plasminogen (Plg), plasmin, urokinase-type plasminogen activator (uPA), its receptor (uPAR), tissue-type plasminogen activator (tPA), α2-antiplasmin (α2AP), and plasminogen activator inhibitor-1 (PAI-1) are expressed in osteoclasts and osteoblasts and are considered essential in maintaining bone homeostasis by regulating the functions of both osteoclasts and osteoblasts. Additionally, fibrinolytic factors are associated with the regulation of inflammation and the immune system. This review explores the roles of fibrinolytic factors in bone destruction caused by inflammation.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Molecular Pathology, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyotanabe, Kyoto 610-0395, Japan
| |
Collapse
|
4
|
Ahmad M, Haffner-Luntzer M, Schoppa A, Najafova Z, Lukic T, Yorgan TA, Amling M, Schinke T, Ignatius A. Mechanical induction of osteoanabolic Wnt1 promotes osteoblast differentiation via Plat. FASEB J 2024; 38:e23489. [PMID: 38407813 DOI: 10.1096/fj.202301424rr] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 01/04/2024] [Accepted: 01/30/2024] [Indexed: 02/27/2024]
Abstract
Physical activity-induced mechanical stimuli play a crucial role in preserving bone mass and structure by promoting bone formation. While the Wnt pathway is pivotal for mediating the osteoblast response to loading, the exact mechanisms are not fully understood. Here, we found that mechanical stimulation induces osteoblastic Wnt1 expression, resulting in an upregulation of key osteogenic marker genes, including Runx2 and Sp7, while Wnt1 knockdown using siRNA prevented these effects. RNAseq analysis identified Plat as a major target through which Wnt1 exerts its osteogenic influence. This was corroborated by Plat depletion using siRNA, confirming its positive role in osteogenic differentiation. Moreover, we demonstrated that mechanical stimulation enhances Plat expression, which, in turn leads to increased expression of osteogenic markers like Runx2 and Sp7. Notably, Plat depletion by siRNA prevented this effect. We have established that Wnt1 regulates Plat expression by activating β-Catenin. Silencing Wnt1 impairs mechanically induced β-Catenin activation, subsequently reducing Plat expression. Furthermore, our findings showed that Wnt1 is essential for osteoblasts to respond to mechanical stimulation and induce Runx2 and Sp7 expression, in part through the Wnt1/β-Catenin/Plat signaling pathway. Additionally, we observed significantly reduced Wnt1 and Plat expression in bones from ovariectomy (OVX)-induced and age-related osteoporotic mouse models compared with non-OVX and young mice, respectively. Overall, our data suggested that Wnt1 and Plat play significant roles in mechanically induced osteogenesis. Their decreased expression in bones from OVX and aged mice highlights their potential involvement in post-menopausal and age-related osteoporosis, respectively.
Collapse
Affiliation(s)
- Mubashir Ahmad
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Melanie Haffner-Luntzer
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | - Astrid Schoppa
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| | | | - Teodora Lukic
- Robert Bosch Center for Tumor Diseases, Stuttgart, Germany
| | - Timur Alexander Yorgan
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Amling
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thorsten Schinke
- Department of Osteology and Biomechanics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Anita Ignatius
- Institute of Orthopedic Research and Biomechanics, University Medical Center Ulm, Ulm, Germany
| |
Collapse
|
5
|
Karnik SJ, Nazzal MK, Kacena MA, Bruzzaniti A. Megakaryocyte Secreted Factors Regulate Bone Marrow Niche Cells During Skeletal Homeostasis, Aging, and Disease. Calcif Tissue Int 2023; 113:83-95. [PMID: 37243755 PMCID: PMC11179715 DOI: 10.1007/s00223-023-01095-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/01/2023] [Indexed: 05/29/2023]
Abstract
The bone marrow microenvironment contains a diverse array of cell types under extensive regulatory control and provides for a novel and complex mechanism for bone regulation. Megakaryocytes (MKs) are one such cell type that potentially acts as a master regulator of the bone marrow microenvironment due to its effects on hematopoiesis, osteoblastogenesis, and osteoclastogenesis. While several of these processes are induced/inhibited through MK secreted factors, others are primarily regulated by direct cell-cell contact. Notably, the regulatory effects that MKs exert on these different cell populations has been found to change with aging and disease states. Overall, MKs are a critical component of the bone marrow that should be considered when examining regulation of the skeletal microenvironment. An increased understanding of the role of MKs in these physiological processes may provide insight into novel therapies that can be used to target specific pathways important in hematopoietic and skeletal disorders.
Collapse
Affiliation(s)
- Sonali J Karnik
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Murad K Nazzal
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Melissa A Kacena
- Department of Orthopaedic Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.
- Richard L. Roudebush VA Medical Center, Indianapolis, IN, USA.
| | - Angela Bruzzaniti
- Department of Biomedical Sciences and Comprehensive Care, Indiana University School of Dentistry, Indianapolis, IN, USA.
| |
Collapse
|
6
|
Brook N, Dharmarajan A, Chan A, Dass CR. Potential therapeutic role for pigment epithelium-derived factor in post-menopausal breast cancer bone metastasis. J Pharm Pharmacol 2023:7146711. [PMID: 37116213 DOI: 10.1093/jpp/rgad039] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
OBJECTIVES This review discusses key oestrogens associated with the circulating pre- and post-menopausal milieu and how they may impact intratumoral oestrogen levels and breast cancer (BC) metastasis. It also identifies critical steps in BC metastasis to bone from the viewpoint of pigment epithelium-derived factor (PEDF) function, and discusses the role of several associated pro-metastatic biomarkers in BC bone metastasis. KEY FINDINGS PEDF is regulated by oestrogen in a number of oestrogen-sensitive tissues. Changes in circulating oestrogen levels associated with menopause may enhance the growth of BC bone metastases, leading to the establishment of a pre-metastatic niche. The establishment of such a pre-metastatic niche is driven by several key mediators, with pro-osteoclastic and pro-metastatic function which are upregulated by BC cells. These mediators appear to be regulated by oestrogen, as well as differentially affected by menopausal status. PEDF interacts with several pro-metastatic, pro-osteoclastic biomarkers, including C-X-C motif chemokine receptor 4 (CXCR4) and nuclear factor kappa B (NFκB) in BC bone metastasis. CONCLUSION Mediators such as CXCR4 and MT1-MMP underpin the ability of PEDF to function as an antimetastatic in other cancers such as osteosarcoma, highlighting the possibility that this serpin could be used as a therapeutic against BC metastasis in future.
Collapse
Affiliation(s)
- Naomi Brook
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| | - Arun Dharmarajan
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai 600116, India
| | - Arlene Chan
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Breast Cancer Research Centre-Western Australia, Hollywood Private Hospital, Nedlands 6009, Australia
| | - Crispin R Dass
- Curtin Medical School, Curtin University, Bentley 6102, Australia
- Curtin Health Innovation Research Institute, Bentley 6102, Australia
| |
Collapse
|
7
|
NFκB-Mediated Mechanisms Drive PEDF Expression and Function in Pre- and Post-Menopausal Oestrogen Levels in Breast Cancer. Int J Mol Sci 2022; 23:ijms232415641. [PMID: 36555293 PMCID: PMC9779285 DOI: 10.3390/ijms232415641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/14/2022] Open
Abstract
Pigment epithelium-derived factor (PEDF) protein regulates normal bone, with anti-tumour roles in bone and breast cancer (BC). Pre- and post-menopausal oestrogen levels may regulate PEDF expression and function in BC, though the mechanisms behind this remain unknown. In this study, in vitro models simulating pre- and post-menopausal bone microenvironments were used to evaluate if PEDF regulates pro-metastatic biomarker expression and downstream functional effects on BC cells. PEDF treatment reduced phosphorylated-nuclear factor-κB p65 subunit (p-NFκB-p65), tumour necrosis factor-α (TNFα), C-X-C chemokine receptor type-4 (CXCR4), and urokinase plasminogen activator receptor (uPAR) in oestrogen receptor (ER)+/human epidermal growth factor receptor-2 (HER2)- BC cells under post-menopausal oestrogen conditions. In triple negative BC (TNBC) cells, PEDF treatment reduced pNFκB-p65 and uPAR expression under pre-menopausal oestrogen conditions. A potential reciprocal regulatory axis between p-NFκB-65 and PEDF in BC was identified, which was BC subtype-specific and differentially regulated by menopausal oestrogen conditions. The effects of PEDF treatment and NFκB inhibition on BC cell function under menopausal conditions were also compared. PEDF treatment exhibited superior anti-viability effects, while combined PEDF and NFκB-p65 inhibitor treatment was superior in reducing BC cell colony formation in a subtype-specific manner. Lastly, immunohistochemical evaluation of p-NFκB-p65 and PEDF expression in human BC and bone metastases specimens revealed an inverse correlation between nuclear PEDF and NFκB expression in bone metastases. We propose that menopausal status is associated with a PEDF/NFκB reciprocal regulatory axis, which drives PEDF expression and anti-metastatic function in a subtype-specific manner. Altogether, our findings identify pre-menopausal TNBC and post-menopausal ER+/HER2- BC patients as target populations for future PEDF research.
Collapse
|
8
|
Tiedemann K, Tsao S, Komarova SV. Platelets and osteoblasts: secretome connections. Am J Physiol Cell Physiol 2022; 323:C347-C353. [PMID: 35675640 DOI: 10.1152/ajpcell.00187.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Megakaryocyte hyperplasia associated with myeloproliferative neoplasms commonly leads to abnormal bone tissue deposition in the bone marrow, known as osteosclerosis. In this study, we aimed to synthesize the known proteomics literature describing factors released by megakaryocytes and platelets and to examine if any of the secreted factors have a known ability to stimulate the bone-forming cells, osteoblasts. Using a systematic search of Medline, we identified 77 articles reporting on factors secreted by platelets and megakaryocytes. After a full-text screening and analysis of the studies, we selected seven papers that reported proteomics data for factors secreted by platelets from healthy individuals. From 60 proteins reported in at least two studies, we focused on 23 that contained a putative signal peptide, which we searched for a potential osteoblast-stimulatory function. From nine proteins with a positive effect on osteoblast formation and function, two extracellular matrix (ECM) proteins, secreted protein acidic and rich in cysteine (SPARC) and tissue inhibitor of metalloproteinase-1 (TIMP1), and three cellular proteins with known extracellular function, the 70-kDa heat shock protein (HSP70), thymosin-β4 (TB4), and super dismutase (SOD), were identified as hypothetical candidate molecules to be examined as potential mediators in mouse models of osteomyelofibrosis. Thus, careful analysis of prior literature can be beneficial in assisting the planning of future experimental studies.
Collapse
Affiliation(s)
- Kerstin Tiedemann
- Faculty of Dental Medicine and Oral Health Sciences, Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| | - Serena Tsao
- Faculty of Dental Medicine and Oral Health Sciences, Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| | - Svetlana V Komarova
- Faculty of Dental Medicine and Oral Health Sciences, Shriners Hospital for Children-Canada, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Wang L, Yao L, Duan H, Yang F, Lin M, Zhang R, He Z, Ahn J, Fan Y, Qin L, Gong Y. Plasminogen Regulates Fracture Repair by Promoting the Functions of Periosteal Mesenchymal Progenitors. J Bone Miner Res 2021; 36:2229-2242. [PMID: 34378815 PMCID: PMC8865375 DOI: 10.1002/jbmr.4423] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 11/06/2022]
Abstract
Defective or insufficient bone repair and regeneration are common in patients as a result of major trauma or severe disease. Cell therapy with periosteal mesenchymal progenitors, which can be limited in severe injury, serves as a promising approach; however, its efficacy is limited due to a repair-hostile ischemic tissue microenvironment after traumatic fracture. Here we report that plasminogen (Plg), a factor that is upregulated in these environments, is critical for fracture healing. Plg knockout mice had impaired trabecular and cortical bone structure and exhibited delayed and incomplete fracture healing. Interestingly, Plg deficiency greatly reduced the thickness of expanded periosteum, suggesting a role of Plg in periosteal mesenchymal progenitor-mediated bone repair. In culture, Plg increased cell proliferation and migration in periosteal mesenchymal progenitors and inhibited cell death under ischemic conditions. Mechanistically, we revealed that Plg cleaved and activated Cyr61 to regulate periosteal progenitor function. Thus, our study uncovers a cellular mechanism underlying fracture healing, by which Plg activates Cyr61 to promote periosteal progenitor proliferation, survival, and migration and improves bone repair after fracture. Targeting Plg may offer a rational and effective therapeutic opportunity for improving fracture healing. © 2021 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Luqiang Wang
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Orthopaedics, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lutian Yao
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Orthopaedics, The First Hospital of China Medical University, Shenyang, China
| | - Hao Duan
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.,Department of Neurosurgery, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Fan Yang
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maohuan Lin
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Rongxin Zhang
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Zhenqiang He
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jaimo Ahn
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yi Fan
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ling Qin
- Departments of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Yanqing Gong
- Division of Translational Medicine and Human Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
10
|
Santibanez JF, Obradović H, Krstić J. BMP2 downregulates urokinase-type plasminogen activator via p38 MAPK: Implications in C2C12 cells myogenic differentiation. Acta Histochem 2021; 123:151774. [PMID: 34450502 DOI: 10.1016/j.acthis.2021.151774] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/05/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Bone morphogenetic protein (BMP)2 strongly affects the differentiation program of myoblast cells by inhibiting myogenesis and inducing osteogenic differentiation. In turn, extracellular matrix (ECM) proteinases, such as urokinase-type plasminogen activator (uPA), can influence the fate of muscle stem cells by participating in ECM reorganization. Although both BMP2 and uPA have antagonistic roles in muscles cells differentiation, no connection between them has been elucidated so far. This study aims to determine whether BMP2 regulates uPA expression in the myogenic C2C12 cell line and its impact on muscle cell fate differentiation. Our results showed that BMP2 did not modify C2C12 cell proliferation in a growth medium or myogenic differentiation medium. Although BMP2 inhibited myogenesis and induced osteogenesis, these effects were achieved with different doses of BMP2. Low concentrations of BMP2 blocked myogenesis, while a higher concentration was needed to induce osteogenesis. Reduced uPA expression was noticed alongside myogenic inhibition at low concentrations of BMP2. BMP2 activated p38 MAPK signaling to inhibit uPA activity. Furthermore, ectopic human uPA expression reduced BMP2's ability to inhibit the myogenic differentiation of C2C12 cells. In conclusion, BMP2 inhibits uPA expression through p38 MAPK and in vitro myogenesis at non-osteogenic concentrations, while uPA ectopic expression prevents BMP2 from inhibiting myogenesis in C2C12 cells.
Collapse
|
11
|
Omidi M, Ahmad Agha N, Müller A, Feyerabend F, Helmholz H, Willumeit-Römer R, Schlüter H, Luthringer-Feyerabend BJC. Investigation of the impact of magnesium versus titanium implants on protein composition in osteoblast by label free quantification. Metallomics 2021; 12:916-934. [PMID: 32352129 DOI: 10.1039/d0mt00028k] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Metallic implant biomaterials predominate in orthopaedic surgery. Compared to titanium-based permanent implants, magnesium-based ones offer new possibilities as they possess mechanical properties closer to the ones of bones and they are biodegradable. Furthermore, magnesium is more and more considered to be "bioactive" i.e., able to elicit a specific tissue response or to strengthen the intimate contact between the implant and the osseous tissue. Indeed, several studies demonstrated the overall beneficial effect of magnesium-based materials on bone tissue (in vivo and in vitro). Here, the direct effects of titanium and magnesium on osteoblasts were measured on proteomes levels in order to highlight metal-specific and relevant proteins. Out of 2100 identified proteins, only 10 and 81 differentially regulated proteins, compare to the control, were isolated for titanium and magnesium samples, respectively. Selected ones according to their relationship to bone tissue were further discussed. Most of them were involved in extracellular matrix maturation and remodelling (two having a negative effect on mineralisation). A fine-tuned balanced between osteoblast maturation, differentiation and viability was observed.
Collapse
Affiliation(s)
- M Omidi
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - N Ahmad Agha
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), 21502 Geesthacht, Germany.
| | - A Müller
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), 21502 Geesthacht, Germany.
| | - F Feyerabend
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), 21502 Geesthacht, Germany.
| | - H Helmholz
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), 21502 Geesthacht, Germany.
| | - R Willumeit-Römer
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), 21502 Geesthacht, Germany.
| | - H Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Centre Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - B J C Luthringer-Feyerabend
- Institute of Materials Research, Division for Metallic Biomaterials, Helmholtz-Zentrum Geesthacht (HZG), 21502 Geesthacht, Germany.
| |
Collapse
|
12
|
Bala K, Cuellar A, Herren AW, Boyadjiev SA. Identification of differentially expressed proteins between fused and open sutures in sagittal nonsyndromic craniosynostosis during suture development by quantitative proteomic analysis. Proteomics Clin Appl 2021; 15:e2000031. [PMID: 33580899 DOI: 10.1002/prca.202000031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 12/09/2020] [Accepted: 12/22/2020] [Indexed: 01/08/2023]
Abstract
PURPOSE Nonsyndromic craniosynostosis (NCS), the premature fusion of cranial sutures, results in an abnormal skull shape and is associated with a significant morbidity. Proteomics is a promising tool for disease characterization and biomarker discovery; we aimed to identify biologically relevant differentially expressed proteins for NCS. EXPERIMENTAL DESIGN Label-based quantitative proteomic profiling using TMT was performed on protein extracted from mesenchymal stem cells, osteoblasts and bone tissue of five open and five fused sutures of sagittal NCS (sNCS) and analyzed using quantitative LC-MS/MS based bottom-up proteomics. Differential protein abundance between open and fused sutures was determined to identify biologically relevant proteins of interest. Proteins were validated in an independent sample set by western blot and immunohistochemistry. RESULTS We observed 838 differentially expressed proteins between open and fused sutures of sNCS. Decorin, lumican, and asporin were significantly downregulated while COL4A1 and TGFβ1|1 were upregulated in fused compared to open sutures. CONCLUSIONS AND CLINICAL RELEVANCE The majority of significantly differentially expressed proteins between open and fused sutures were observed in the proteomes of osteoblasts suggesting that protein changes contributing to premature sagittal suture fusion occur predominantly at the osteoblast level. Our findings suggest a possible ineffective ECM deposition at the osteoblast cell stage.
Collapse
Affiliation(s)
- Krithi Bala
- Department of Pediatrics, University of California Davis, Sacramento, California, USA
| | - Araceli Cuellar
- Department of Pediatrics, University of California Davis, Sacramento, California, USA
| | - Anthony W Herren
- UC Davis Genome Center, University of California Davis, Davis, California, USA
| | - Simeon A Boyadjiev
- Department of Pediatrics, University of California Davis, Sacramento, California, USA
| |
Collapse
|
13
|
Baranowsky A, Appelt J, Tseneva K, Jiang S, Jahn D, Tsitsilonis S, Frosch KH, Keller J. Tranexamic Acid Promotes Murine Bone Marrow-Derived Osteoblast Proliferation and Inhibits Osteoclast Formation In Vitro. Int J Mol Sci 2021; 22:ijms22010449. [PMID: 33466312 PMCID: PMC7795046 DOI: 10.3390/ijms22010449] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 12/28/2020] [Accepted: 12/30/2020] [Indexed: 12/22/2022] Open
Abstract
Despite modern surgical trauma care, bleeding contributes to one-third of trauma-related death. A significant improvement was obtained through the introduction of tranexamic acid (TXA), which today is widely used in emergency and elective orthopedic surgery to control bleeding. However, concerns remain regarding potential adverse effects on bone turnover and regeneration. Therefore, we employed standardized cell culture systems including primary osteoblasts, osteoclasts, and macrophages to evaluate potential effects of TXA on murine bone cells. While osteoblasts derived from calvarial digestion were not affected, TXA increased cell proliferation and matrix mineralization in bone marrow-derived osteoblasts. Short-term TXA treatment (6 h) failed to alter the expression of osteoblast markers; however, long-term TXA stimulation (10 days) was associated with the increased expression of genes involved in osteoblast differentiation and extracellular matrix synthesis. Similarly, whereas short-term TXA treatment did not affect gene expression in terminally differentiated osteoclasts, long-term TXA stimulation resulted in the potent inhibition of osteoclastogenesis. Finally, in bone marrow-derived macrophages activated with LPS, simultaneous TXA treatment led to a reduced expression of inflammatory cytokines and chemokines. Collectively, our study demonstrates a differential action of TXA on bone cells including osteoanabolic, anti-resorptive, and anti-inflammatory effects in vitro which suggests novel treatment applications.
Collapse
Affiliation(s)
- Anke Baranowsky
- Clinic of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (A.B.); (S.J.); (K.-H.F.)
| | - Jessika Appelt
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (J.A.); (K.T.); (D.J.); (S.T.)
| | - Kristina Tseneva
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (J.A.); (K.T.); (D.J.); (S.T.)
| | - Shan Jiang
- Clinic of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (A.B.); (S.J.); (K.-H.F.)
| | - Denise Jahn
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (J.A.); (K.T.); (D.J.); (S.T.)
| | - Serafeim Tsitsilonis
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (J.A.); (K.T.); (D.J.); (S.T.)
- Center for Musculoskeletal Surgery, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Karl-Heinz Frosch
- Clinic of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (A.B.); (S.J.); (K.-H.F.)
| | - Johannes Keller
- Clinic of Trauma and Orthopedic Surgery, University Medical Center Hamburg-Eppendorf, 20251 Hamburg, Germany; (A.B.); (S.J.); (K.-H.F.)
- Correspondence: ; Tel.: +49-(0)40-7410-5-6691
| |
Collapse
|
14
|
Interleukin-17 modulates uPA and MMP2 expression in human periodontal ligament mesenchymal stem cells: Involvement of the ERK1/2 MAPK pathway. ARCH BIOL SCI 2021. [DOI: 10.2298/abs210929048o] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Periodontal disease is a chronic infection of periodontal tissue
characterized by extracellular matrix (ECM) degradation due to increased
expression of plasminogen activators and matrix metalloproteinases (MMPs)
and various proinflammatory cytokines, including interleukin (IL)-17.
Successful regeneration of damaged periodontal tissues depends on the proper
functionality of periodontal ligament mesenchymal stem cells (PDLMSCs),
especially the production of extracellular matrix proteases. We investigated
the influence of IL-17 on ECM remodeling through modulation of urokinasetype
plasminogen activator (uPA) and MMP2/MMP9 expression in human PDLMSCs at
mRNA, protein and activity levels using by RT-PCR, Western blotting and
zymography, respectively. Investigation of the involvement of MAPKs in these
processes in PDLMSCs was determined by Western blotting, as well as by
utilizing specific p38 and MEK1/2 inhibitors. Our results show that IL-17
activates MAPK signaling in PDLMSCs. Moreover, IL-17 had no effect on MMP9
expression, but it stimulated uPA and MMP2 gene and protein expression in
PDLMSCs through the activation of the ERK1/2 MAPK signaling pathway. The
obtained data suggest that IL-17 contributes to ECM degradation in the
periodontal ligament by stimulating uPA and MMP2 expression and activity in
PDLMSCs. This information is important for understanding periodontal disease
development and defining future directions of its treatment.
Collapse
|
15
|
Socorro M, Shinde A, Yamazaki H, Khalid S, Monier D, Beniash E, Napierala D. Trps1 transcription factor represses phosphate-induced expression of SerpinB2 in osteogenic cells. Bone 2020; 141:115673. [PMID: 33022456 PMCID: PMC7680451 DOI: 10.1016/j.bone.2020.115673] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 09/24/2020] [Accepted: 09/29/2020] [Indexed: 12/13/2022]
Abstract
Serine protease inhibitor SerpinB2 is one of the most upregulated proteins following cellular stress. This multifunctional serpin has been attributed a number of pleiotropic activities, including roles in cell survival, proliferation, differentiation, immunity and extracellular matrix (ECM) remodeling. Studies of cancer cells demonstrated that expression of SerpinB2 is directly repressed by the Trps1 transcription factor, which is a regulator of skeletal and dental tissues mineralization. In our previous studies, we identified SerpinB2 as one of the novel genes highly upregulated by phosphate (Pi) at the initiation of the mineralization process, however SerpinB2 has never been implicated in formation nor homeostasis of mineralized tissues. The aim of this study was to establish, if SerpinB2 is involved in function of cells producing mineralized ECM and to determine the interplay between Pi signaling and Trps1 in the regulation of SerpinB2 expression specifically in cells producing mineralized ECM. Analyses of the SerpinB2 expression pattern in mouse skeletal and dental tissues detected high SerpinB2 protein levels specifically in cells producing mineralized ECM. qRT-PCR and Western blot analyses demonstrated that SerpinB2 expression is activated by elevated Pi specifically in osteogenic cells. However, the Pi-induced SerpinB2 expression was diminished by overexpression of Trps1. Decreased SerpinB2 levels were also detected in osteoblasts and odontoblasts of 2.3Col1a1-Trps1 transgenic mice. Chromatin immunoprecipitation assay (ChIP) revealed that the occupancy of Trps1 on regulatory elements in the SerpinB2 gene changes in response to Pi. In vitro functional assessment of the consequences of SerpinB2 deficiency in cells producing mineralized ECM detected impaired mineralization in SerpinB2-deficient cells in comparison with controls. In conclusion, high and specific expression of SerpinB2 in cells producing mineralized ECM, the impaired mineralization of SerpinB2-deficient cells and regulation of SerpinB2 expression by two molecules regulating formation of mineralized tissues suggest involvement of SerpinB2 in physiological mineralization.
Collapse
Affiliation(s)
- Mairobys Socorro
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Apurva Shinde
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Hajime Yamazaki
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Sana Khalid
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Daisy Monier
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA
| | - Elia Beniash
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Dobrawa Napierala
- Center for Craniofacial Regeneration, Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, PA, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
16
|
Okada K, Nishioka M, Kaji H. Roles of fibrinolytic factors in the alterations in bone marrow hematopoietic stem/progenitor cells during bone repair. Inflamm Regen 2020; 40:22. [PMID: 32944096 PMCID: PMC7493393 DOI: 10.1186/s41232-020-00128-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 06/30/2020] [Indexed: 12/31/2022] Open
Abstract
In bone tissues, metabolic turnover through bone resorption by osteoclasts and bone formation by osteoblasts, termed bone remodeling, is strictly controlled and maintains homeostasis. Fibrinolytic factors are expressed in osteoclasts and osteoblasts, and are involved in bone remodeling through bone resorption and formation. The repair/regeneration process after bone injury is divided into the acute inflammatory, repair, and remodeling stages. Osteoblasts, osteoclasts, chondrocytes, and macrophages involved in the bone repair process originate from hematopoietic stem/progenitor cells (HSPCs) and mesenchymal stem cells (MSCs) in the bone marrow. Therefore, stem cells in the bone marrow may be strongly influenced by bone injury. The urokinase-type PA (u-PA)/plasminogen (Plg) system functions in macrophage accumulation/phagocytosis through chemokines in the acute inflammatory stage, and Plg increases blood vessel-related growth factor expression, being involved in vascularization in mice. Plasminogen activator inhivitor-1 (PAI-1) causes bone loss and delayed bone repair through the inhibition of osteoblast differentiation in a drug-induced diabetes model in mice. Plg is considered to induce transforming growth factor-β (TGF-β) production in macrophages in the bone repair process, TGF-β release from the extracellular matrix through the activation of matrix metalloproteinase-9 (MMP-9), and stromal cell-derived factor-1 (SDF-1) expression in endosteal preosteoblasts, leading to the induction of bone marrow HSPCs in mice. Based on the above, establishment of a fibrinolytic factor-targeting method efficiently promoting bone repair/regeneration and fracture healing, and development of a new osteoporosis treatment method and diagnostic marker are awaited.
Collapse
Affiliation(s)
- Kiyotaka Okada
- Department of Arts and Science, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, 589-8511 Japan.,Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, 589-8511 Japan
| | - Minoru Nishioka
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, 589-8511 Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kindai University Faculty of Medicine, Osaka-Sayama, Osaka, 589-8511 Japan
| |
Collapse
|
17
|
Bravo D, Josephson AM, Bradaschia-Correa V, Wong MZ, Yim NL, Neibart SS, Lee SN, Huo J, Coughlin T, Mizrahi MM, Leucht P. Temporary inhibition of the plasminogen activator inhibits periosteal chondrogenesis and promotes periosteal osteogenesis during appendicular bone fracture healing. Bone 2018; 112:97-106. [PMID: 29680264 PMCID: PMC5970081 DOI: 10.1016/j.bone.2018.04.016] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Revised: 02/11/2018] [Accepted: 04/17/2018] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Aminocaproic acid is approved as an anti-fibrinolytic for use in joint replacement and spinal fusion surgeries to limit perioperative blood loss. Previous animal studies have demonstrated a pro-osteogenic effect of aminocaproic acid in spine fusion models. Here, we tested if aminocaproic acid enhances appendicular bone healing and we sought to uncover the effect of aminocaproic acid on osteoprogenitor cells (OPCs) during bone regeneration. METHODS We employed a well-established murine femur fracture model in adult C57BL/6J mice after receiving two peri-operative injections of aminocaproic acid. Routine histological assays, biomechanical testing and micro-CT analyses were utilized to assess callus volume, and strength, progenitor cell proliferation, differentiation, and remodeling in vivo. Two disparate ectopic transplantation models were used to study the effect of the growth factor milieu within the early fracture hematoma on osteoprogenitor cell fate decisions. RESULTS Aminocaproic acid treated femur fractures healed with a significantly smaller cartilaginous callus, and this effect was also observed in the ectopic transplantation assays. We hypothesized that aminocaproic acid treatment resulted in a stabilization of the early fracture hematoma, leading to a change in the growth factor milieu created by the early hematoma. Gene and protein expression analysis confirmed that aminocaproic acid treatment resulted in an increase in Wnt and BMP signaling and a decrease in TGF-β-signaling, resulting in a shift from chondrogenic to osteogenic differentiation in this model of endochondral bone formation. CONCLUSION These experiments demonstrate for the first time that inhibition of the plasminogen activator during fracture healing using aminocaproic acid leads to a change in cell fate decision of periosteal osteoprogenitor cells, with a predominance of osteogenic differentiation, resulting in a larger and stronger bony callus. These findings may offer a promising new use of aminocaproic acid, which is already FDA-approved and offers a very safe risk profile.
Collapse
Affiliation(s)
- D Bravo
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - A M Josephson
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - V Bradaschia-Correa
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - M Z Wong
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - N L Yim
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - S S Neibart
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - S N Lee
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - J Huo
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - T Coughlin
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - M M Mizrahi
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States
| | - P Leucht
- Department of Orthopaedic Surgery, New York University School of Medicine, New York, NY, United States; Department of Cell Biology, New York University School of Medicine, New York, NY, United States.
| |
Collapse
|
18
|
Bayraktar S, Jungbluth P, Deenen R, Grassmann J, Schneppendahl J, Eschbach D, Scholz A, Windolf J, Suschek CV, Grotheer V. Molecular- and microarray-based analysis of diversity among resting and osteogenically induced porcine mesenchymal stromal cells of several tissue origin. J Tissue Eng Regen Med 2017; 12:114-128. [PMID: 27966263 PMCID: PMC5811815 DOI: 10.1002/term.2375] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 11/12/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022]
Abstract
Mesenchymal stromal cells (MSCs) play a pivotal role in modern therapeutic approaches in bone‐healing disorders. Although bone marrow‐derived MSCs are most frequently used, the knowledge that many other adult tissues represent promising sources for potent MSCs has gained acceptance. In the present study, the osteogenic differentiation potential of porcine skin fibroblasts (FBs), as well as bone marrow‐ (BMSCs), adipose tissue‐ (ASCs) and dental pulp‐derived stromal cells (DSCs) were evaluated. However, additional application of BMP‐2 significantly elevated the delayed osteogenic differentiation capacity of ASC and FB cultures, and in DSC cultures the supplementation of platelet‐rich plasma increased osteogenic differentiation potential to a comparable level of the good differentiable BMSCs. Furthermore, microarray gene expression performed in an exemplary manner for ASCs and BMSCs revealed that ASCs and BMSCs use different gene expression patterns for osteogenic differentiation under standard media conditions, as diverse MSCs are imprinted dependent from their tissue niche. However, after increasing the differentiation potential of ASCs to a comparable level as shown in BMSCs, a small subset of identical key molecules was used to differentiate in the osteogenic lineage. Until now, the importance of identified genes seems to be underestimated for osteogenic differentiation. Apparently, the regulation of transmembrane protein 229A, interleukin‐33 and the fibroblast growth factor receptor‐2 in the early phase of osteogenic differentiation is needed for optimum results. Based on these results, bone regeneration strategies of MSCs have to be adjusted, and in vivo studies on the osteogenic capacities of the different types of MCSs are warranted. Copyright © 2016 The Authors Tissue Engineering and Regenerative Medicine published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Samet Bayraktar
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Pascal Jungbluth
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - René Deenen
- Biological and Medical Research Center (BMFZ), Genomics and Transcriptomics Laboratory (GTL), Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Jan Grassmann
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Johannes Schneppendahl
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Daphne Eschbach
- Department of Trauma-, Hand- and Reconstructive Surgery, University of Giessen and Marburg, Location Marburg, 35033, Marburg, Germany
| | - Armin Scholz
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Joachim Windolf
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Christoph V Suschek
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Vera Grotheer
- Department of Trauma and Hand Surgery, Medical Faculty, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| |
Collapse
|
19
|
Ciavarella S, Laurenzana A, De Summa S, Pilato B, Chillà A, Lacalamita R, Minoia C, Margheri F, Iacobazzi A, Rana A, Merchionne F, Fibbi G, Del Rosso M, Guarini A, Tommasi S, Serratì S. u-PAR expression in cancer associated fibroblast: new acquisitions in multiple myeloma progression. BMC Cancer 2017; 17:215. [PMID: 28340565 PMCID: PMC5366111 DOI: 10.1186/s12885-017-3183-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 03/09/2017] [Indexed: 01/07/2023] Open
Abstract
Background Multiple Myeloma (MM) is a B-cell malignancy in which clonal plasma cells progressively expand within the bone marrow (BM) as effect of complex interactions with extracellular matrix and a number of microenvironmental cells. Among these, cancer-associated fibroblasts (CAF) mediate crucial reciprocal signals with MM cells and are associated to aggressive disease and poor prognosis. A large body of evidence emphasizes the role of the urokinase plasminogen activator (u-PA) and its receptor u-PAR in potentiating the invasion capacity of tumor plasma cells, but little is known about their role in the biology of MM CAF. In this study, we investigated the u-PA/u-PAR axis in MM-associated fibroblasts and explore additional mechanisms of tumor/stroma interplay in MM progression. Methods CAF were purified from total BM stromal fraction of 64 patients including monoclonal gammopathy of undetermined significance, asymptomatic and symptomatic MM, as well as MM in post-treatment remission. Flow cytometry, Real Time PCR and immunofluorescence were performed to investigate the u-PA/u-PAR system in relation to the level of activation of CAF at different stages of the disease. Moreover, proliferation and invasion assays coupled with silencing experiments were used to prove, at functional level, the function of u-PAR in CAF. Results We found higher activation level, along with increased expression of pro-invasive molecules, including u-PA, u-PAR and metalloproteinases, in CAF from patients with symptomatic MM compared to the others stages of the disease. Consistently, CAF from active MM as well as U266 cell line under the influence of medium conditioned by active MM CAF, display higher proliferative rate and invasion potential, which were significantly restrained by u-PAR gene expression inhibition. Conclusions Our data suggest that the stimulation of u-PA/u-PAR system contributes to the activated phenotype and function of CAF during MM progression, providing a biological rationale for future targeted therapies against MM.
Collapse
Affiliation(s)
- S Ciavarella
- National Cancer Research Centre IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - A Laurenzana
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - S De Summa
- Molecular Genetics Laboratory, National Cancer Research Centre, IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - B Pilato
- Molecular Genetics Laboratory, National Cancer Research Centre, IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - A Chillà
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - R Lacalamita
- Molecular Genetics Laboratory, National Cancer Research Centre, IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - C Minoia
- National Cancer Research Centre IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - F Margheri
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - A Iacobazzi
- National Cancer Research Centre IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - A Rana
- National Cancer Research Centre IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - F Merchionne
- National Cancer Research Centre IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - G Fibbi
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - M Del Rosso
- Department of Experimental and Clinical Biomedical Sciences, Section of Experimental Pathology and Oncology, University of Florence, Florence, Italy
| | - A Guarini
- National Cancer Research Centre IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - S Tommasi
- Molecular Genetics Laboratory, National Cancer Research Centre, IRCCS "Giovanni Paolo II", 70124, Bari, Italy
| | - S Serratì
- Molecular Genetics Laboratory, National Cancer Research Centre, IRCCS "Giovanni Paolo II", 70124, Bari, Italy. .,Nanotecnology Laboratory, National Cancer Research Centre, IRCCS "Giovanni Paolo II", Bari, Italy.
| |
Collapse
|
20
|
Romero-Gavilán F, Gomes NC, Ródenas J, Sánchez A, Azkargorta M, Iloro I, Elortza F, García Arnáez I, Gurruchaga M, Goñi I, Suay J. Proteome analysis of human serum proteins adsorbed onto different titanium surfaces used in dental implants. BIOFOULING 2017; 33:98-111. [PMID: 28005415 DOI: 10.1080/08927014.2016.1259414] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Accepted: 10/31/2016] [Indexed: 06/06/2023]
Abstract
Titanium dental implants are commonly used due to their biocompatibility and biochemical properties; blasted acid-etched Ti is used more frequently than smooth Ti surfaces. In this study, physico-chemical characterisation revealed important differences in roughness, chemical composition and hydrophilicity, but no differences were found in cellular in vitro studies (proliferation and mineralization). However, the deposition of proteins onto the implant surface might affect in vivo osseointegration. To test that hypothesis, protein layers formed on discs of both surface type after incubation with human serum were analysed. Using mass spectrometry (LC/MS/MS), 218 proteins were identified, 30 of which were associated with bone metabolism. Interestingly, Apo E, antithrombin and protein C adsorbed mostly onto blasted and acid-etched Ti, whereas the proteins of the complement system (C3) were found predominantly on smooth Ti surfaces. These results suggest that physico-chemical characteristics could be responsible for the differences observed in the adsorbed protein layer.
Collapse
Affiliation(s)
- Francisco Romero-Gavilán
- a Department of Industrial Systems and Design Engineering , University of Castellón , Castellón de la Plana , Spain
| | - N C Gomes
- b Department of Medicine , University of Castellón , Castellón de la Plana , Spain
| | - Joaquin Ródenas
- a Department of Industrial Systems and Design Engineering , University of Castellón , Castellón de la Plana , Spain
| | - Ana Sánchez
- b Department of Medicine , University of Castellón , Castellón de la Plana , Spain
| | - Mikel Azkargorta
- c Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed-ISCIII , Derio , Spain
| | - Ibon Iloro
- c Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed-ISCIII , Derio , Spain
| | - Felix Elortza
- c Proteomics Platform, CIC bioGUNE, CIBERehd, ProteoRed-ISCIII , Derio , Spain
| | - Iñaki García Arnáez
- d Department of Polymer Science and Technology , University of Basque Country , San Sebastián , Spain
| | - Mariló Gurruchaga
- d Department of Polymer Science and Technology , University of Basque Country , San Sebastián , Spain
| | - Isabel Goñi
- d Department of Polymer Science and Technology , University of Basque Country , San Sebastián , Spain
| | - Julio Suay
- a Department of Industrial Systems and Design Engineering , University of Castellón , Castellón de la Plana , Spain
| |
Collapse
|
21
|
Kim JO, Han SH, Lee YH, Ahn TK, Lim JJ, Chung YS, Shin DE, Lee WS, Han IB, Kim NK. Association of Plasminogen Activator Inhibitor-1 (PAI-1) Gene Polymorphisms with Osteoporotic Vertebral Compression Fractures (OVCFs) in Postmenopausal Women. Int J Mol Sci 2016; 17:E2062. [PMID: 27941685 PMCID: PMC5187862 DOI: 10.3390/ijms17122062] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Revised: 12/02/2016] [Accepted: 12/04/2016] [Indexed: 01/01/2023] Open
Abstract
Osteoporosis and osteoporotic fractures are strongly associated with mortality and morbidity, both in developing and developed countries. Menopause accelerates bone loss due to estrogen deficiency and age-related linear bone loss. We investigated plasminogen activator inhibitor-1 (PAI-1) gene polymorphisms in postmenopausal women with osteoporotic vertebral compression fractures (OVCFs). In this case-control study, 355 postmenopausal women were genotyped for the presence of PAI-1 gene polymorphisms -844A > G, -675 4G > 5G, 43G > A, 9785A > G, and 11053T > G. Genetic polymorphisms of PAI-1 were analyzed by the polymerization chain reaction restriction fragment length polymorphism assay, and their association with disease status and folate and homocysteine levels was determined in 158 OVCF patients and 197 control subjects. The PAI-1 -675 5G5G (adjusted odds ratio (AOR), 3.302; p = 0.017) and 43GA + AA (AOR, 2.087; p = 0.042) genotype frequencies showed significant association with the increased prevalence of OVCFs in postmenopausal women. In addition, we performed gene-environment interaction studies and demonstrated an association between PAI-1 gene polymorphisms and OVCF prevalence. Our novel finding is the identification of several PAI-1 genetic variants that increase susceptibility to OVCF. Our findings suggest that polymorphisms in PAI-1 may contribute to OVCF, and that they can be developed as biomarkers for evaluating OVCF risk.
Collapse
Affiliation(s)
- Jung Oh Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea.
| | - Soo Hong Han
- Department of Orthopedics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea.
| | - Yeon Ho Lee
- Department of Orthopedics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea.
| | - Tae Keun Ahn
- Department of Orthopedics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea.
| | - Jae Joon Lim
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea.
| | - Young Sun Chung
- Department of Neurosurgery, Konkuk University Chungju Hospital, Chungju 27376, Korea.
| | - Dong Eun Shin
- Department of Orthopedics, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea.
| | - Woo Sik Lee
- Fertility Center of CHA Gangnam Medical Center, CHA University, Seoul 06135, Korea.
| | - In Bo Han
- Department of Neurosurgery, CHA Bundang Medical Center, CHA University, Seongnam 13496, Korea.
| | - Nam Keun Kim
- Department of Biomedical Science, College of Life Science, CHA University, Seongnam 13488, Korea.
| |
Collapse
|
22
|
Wehner C, Janjić K, Agis H. Relevance of the plasminogen system in physiology, pathology, and regeneration of oral tissues - From the perspective of dental specialties. Arch Oral Biol 2016; 74:136-145. [PMID: 27743595 DOI: 10.1016/j.archoralbio.2016.09.014] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 09/15/2016] [Accepted: 09/30/2016] [Indexed: 12/15/2022]
Abstract
Plasmin is a proteolytic enzyme that is crucial in fibrinolysis. In oral tissues, the plasminogen system plays an essential role in physiological and pathological processes, which in addition to fibrinolysis include degradation of extracellular matrix, inflammation, immune response, angiogenesis, tissue remodeling, cell migration, and wound healing. Oral tissues reveal a change in the plasminogen system during pathological processes such as periodontitis, peri-implantitis, or pulpitis, as well as in response to mechanical load. The plasminogen system is also a key element in tissue regeneration. The number of studies investigating the plasminogen system in dentistry have grown continuously in recent years, highlighting its increasing relevance in dental medicine. In this review, we present the diverse functions of the plasminogen system in physiology and its importance for dental specialists in pathology and regeneration. We thus provide an overview of the current knowledge on the role of the plasminogen system in the different fields of dentistry, including endodontics, orthodontics, periodontics, and oral surgery.
Collapse
Affiliation(s)
- Christian Wehner
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University of Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Klara Janjić
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University of Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Hermann Agis
- Department of Conservative Dentistry and Periodontology, School of Dentistry, Medical University of Vienna, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
23
|
Kaji H. Adipose Tissue‐Derived Plasminogen Activator Inhibitor‐1 Function and Regulation. Compr Physiol 2016; 6:1873-1896. [DOI: 10.1002/cphy.c160004] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Fleetwood AJ, O'Brien-Simpson NM, Veith PD, Lam RS, Achuthan A, Cook AD, Singleton W, Lund IK, Reynolds EC, Hamilton JA. Porphyromonas gingivalis-derived RgpA-Kgp Complex Activates the Macrophage Urokinase Plasminogen Activator System: IMPLICATIONS FOR PERIODONTITIS. J Biol Chem 2015; 290:16031-42. [PMID: 25979345 PMCID: PMC4481207 DOI: 10.1074/jbc.m115.645572] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/12/2015] [Indexed: 01/07/2023] Open
Abstract
Urokinase plasminogen activator (uPA) converts plasminogen to plasmin, resulting in a proteolytic cascade that has been implicated in tissue destruction during inflammation. Periodontitis is a highly prevalent chronic inflammatory disease characterized by destruction of the tissue and bone that support the teeth. We demonstrate that stimulation of macrophages with the arginine- and lysine-specific cysteine protease complex (RgpA-Kgp complex), produced by the keystone pathogen Porphyromonas gingivalis, dramatically increased their ability to degrade matrix in a uPA-dependent manner. We show that the RgpA-Kgp complex cleaves the inactive zymogens, pro-uPA (at consensus sites Lys(158)-Ile(159) and Lys(135)-Lys(136)) and plasminogen, yielding active uPA and plasmin, respectively. These findings are consistent with activation of the uPA proteolytic cascade by P. gingivalis being required for the pathogen to induce alveolar bone loss in a model of periodontitis and reveal a new host-pathogen interaction in which P. gingivalis activates a critical host proteolytic pathway to promote tissue destruction and pathogen virulence.
Collapse
Affiliation(s)
- Andrew J Fleetwood
- From the Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia,
| | - Neil M O'Brien-Simpson
- the Oral Health Cooperative Research Centre, Melbourne Dental School, University of Melbourne, Victoria 3010, Australia, and
| | - Paul D Veith
- the Oral Health Cooperative Research Centre, Melbourne Dental School, University of Melbourne, Victoria 3010, Australia, and
| | - Roselind S Lam
- the Oral Health Cooperative Research Centre, Melbourne Dental School, University of Melbourne, Victoria 3010, Australia, and
| | - Adrian Achuthan
- From the Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - Andrew D Cook
- From the Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| | - William Singleton
- the Oral Health Cooperative Research Centre, Melbourne Dental School, University of Melbourne, Victoria 3010, Australia, and
| | - Ida K Lund
- the Finsen Laboratory, Rigshospitalet and the Biotech Research and Innovation Centre, Copenhagen University, 1165 Copenhagen, Denmark
| | - Eric C Reynolds
- the Oral Health Cooperative Research Centre, Melbourne Dental School, University of Melbourne, Victoria 3010, Australia, and
| | - John A Hamilton
- From the Department of Medicine, University of Melbourne, Royal Melbourne Hospital, Parkville, Victoria 3050, Australia
| |
Collapse
|
25
|
The Tissue Fibrinolytic System Contributes to the Induction of Macrophage Function and CCL3 during Bone Repair in Mice. PLoS One 2015; 10:e0123982. [PMID: 25893677 PMCID: PMC4404328 DOI: 10.1371/journal.pone.0123982] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2014] [Accepted: 02/24/2015] [Indexed: 11/19/2022] Open
Abstract
Macrophages play crucial roles in repair process of various tissues. However, the details in the role of macrophages during bone repair still remains unknown. Herein, we examined the contribution of the tissue fibrinolytic system to the macrophage functions in bone repair after femoral bone defect by using male mice deficient in plasminogen (Plg–/–), urokinase-type plasminogen activator (uPA–/–) or tissue-type plasminogen activator (tPA–/–) genes and their wild-type littermates. Bone repair of the femur was delayed in uPA–/– mice until day 6, compared with wild-type (uPA+/+) mice. Number of Osterix-positive cells and vessel formation were decreased in uPA–/– mice at the bone injury site on day 4, compared with those in uPA+/+ mice. Number of macrophages and their phagocytosis at the bone injury site were reduced in uPA–/– and Plg–/–, but not in tPA–/– mice on day 4. Although uPA or plasminogen deficiency did not affect the levels of cytokines, including TNF-α, IL-1β, IL-6, IL-4 and IFN-γ mRNA in the damaged femur, the elevation in CCL3 mRNA levels was suppressed in uPA–/– and Plg–/–, but not in tPA–/– mice. Neutralization of CCL3 antagonized macrophage recruitment to the site of bone injury and delayed bone repair in uPA+/+, but not in uPA–/– mice. Our results provide novel evidence that the tissue fibrinolytic system contributes to the induction of macrophage recruitment and CCL3 at the bone injury site, thereby, leading to the enhancement of the repair process.
Collapse
|
26
|
Tudpor K, van der Eerden BCJ, Jongwattanapisan P, Roelofs JJTH, van Leeuwen JPTM, Bindels RJM, Hoenderop JGJ. Thrombin receptor deficiency leads to a high bone mass phenotype by decreasing the RANKL/OPG ratio. Bone 2015; 72:14-22. [PMID: 25460576 DOI: 10.1016/j.bone.2014.11.004] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 10/26/2014] [Accepted: 11/10/2014] [Indexed: 12/25/2022]
Abstract
Thrombin and its receptor (TR) are, respectively, expressed in osteoclasts and osteoblasts. However, their physiological roles on bone metabolism have not been fully elucidated. Here we investigated the bone microarchitecture by micro-computed tomography (μCT) and demonstrated increased trabecular and cortical bone mass in femurs of TR KO mice compared to WT littermates. Trabecular thickness and connectivity were significantly enhanced. The physiological role of TR on both inorganic and organic phases of bone is illustrated by a significant increase in BMD and a decrease in urinary deoxypyridinoline (DPD) crosslink concentration in TR KO mice. Moreover, TR KO cortical bone expanded and had a higher polar moment of inertia (J), implying stronger bone. Bone histomorphometry illustrated unaltered osteoblast and osteoclast number and surface in femoral metaphyses, indicating that thrombin/TR regulates osteoblasts and osteoclasts at functional levels. Serum analysis showed a decrease in RANKL and an increase in osteoprotegerin (OPG) levels and reflected a reduced RANKL/OPG ratio in the TR KO group. In vitro experiments using MC3T3 pre-osteoblasts demonstrated a TR-dependent stimulatory effect of thrombin on the RANKL/OPG ratio. This effect was blocked by TR antagonist and p42/p44-ERK inhibitor. In addition, thrombin also intensified p42/p44-ERK expression and phosphorylation. In conclusion, the thrombin/TR system maintains normal bone remodeling by activating RANKL and limiting OPG synthesis by osteoblasts through the p42/44-ERK signaling pathway. Consequently, TR deficiency inhibits osteoclastogenesis, resulting in a high bone mass phenotype.
Collapse
Affiliation(s)
- Kukiat Tudpor
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands
| | | | - Prapaporn Jongwattanapisan
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands
| | - Joris J T H Roelofs
- Department of Pathology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - René J M Bindels
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands
| | - Joost G J Hoenderop
- Department of Physiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, The Netherlands.
| |
Collapse
|
27
|
Rantala J, Kemppainen S, Ndode-Ekane XE, Lahtinen L, Bolkvadze T, Gurevicius K, Tanila H, Pitkänen A. Urokinase-type plasminogen activator deficiency has little effect on seizure susceptibility and acquired epilepsy phenotype but reduces spontaneous exploration in mice. Epilepsy Behav 2015; 42:117-28. [PMID: 25506794 DOI: 10.1016/j.yebeh.2014.11.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2014] [Revised: 11/03/2014] [Accepted: 11/08/2014] [Indexed: 01/05/2023]
Abstract
Urokinase-type plasminogen activator (uPA), a serine protease, converts plasminogen to plasmin. Activation of plasmin leads to degradation of the extracellular matrix, which is critical for tissue recovery, angiogenesis, cell migration, and axonal and synaptic plasticity. We hypothesized that uPA deficiency would cause an abnormal neurophenotype and would lead to exacerbated epileptogenesis after brain injury. Wild-type (Wt) and uPA-/- mice underwent a battery of neurologic behavioral tests evaluating general reactivity, spontaneous exploratory activity, motor coordination, pain threshold, fear and anxiety, and memory. We placed particular emphasis on the effect of uPA deficiency on seizure susceptibility, including the response to convulsants (pentylenetetrazol, kainate, or pilocarpine) and kainate-induced epileptogenesis and epilepsy. The uPA-/- mice showed no motor or sensory impairment compared with the Wt mice. Hippocampus-dependent spatial memory also remained intact. The uPA-/- mice, however, exhibited reduced exploratory activity and an enhanced response to a tone stimulus (p<0.05 compared with the Wt mice). The urokinase-type plasminogen activator deficient mice showed no increase in spontaneous or evoked epileptiform electrographic activity. Rather, the response to pilocarpine administration was reduced compared with the Wt mice (p<0.05). Also, the epileptogenesis and the epilepsy phenotype after intrahippocampal kainate injection were similar to those in the Wt mice. Taken together, uPA deficiency led to diminished interest in the environmental surroundings and enhanced emotional reactivity to unexpected aversive stimuli. Urokinase-type plasminogen activator deficiency was not associated with enhanced seizure susceptibility or worsened poststatus epilepticus epilepsy phenotype.
Collapse
Affiliation(s)
- J Rantala
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - S Kemppainen
- Neurobiology of Memory Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - X E Ndode-Ekane
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - L Lahtinen
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - Tamuna Bolkvadze
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - K Gurevicius
- Neurobiology of Memory Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland
| | - H Tanila
- Neurobiology of Memory Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, PO Box 1777, FIN-70211 Kuopio, Finland
| | - A Pitkänen
- Epilepsy Research Laboratory, A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, PO Box 1627, FIN-70211 Kuopio, Finland; Department of Neurology, Kuopio University Hospital, PO Box 1777, FIN-70211 Kuopio, Finland.
| |
Collapse
|
28
|
Tananuvat N, Charoenkwan P, Ohazama A, Ketuda Cairns JR, Kaewgahya M, Kantaputra PN. Root dentin anomaly and a PLG mutation. Eur J Med Genet 2014; 57:630-5. [DOI: 10.1016/j.ejmg.2014.09.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 09/21/2014] [Indexed: 10/24/2022]
|
29
|
Abstract
STUDY DESIGN Animal model. OBJECTIVE To determine whether aminocaproic acid (Amicar) and tranexamic acid (TXA) inhibit spine fusion volume. SUMMARY OF BACKGROUND DATA Amicar and TXA are antifibrinolytics used to reduce perioperative bleeding. Prior in vitro data showed that antifibrinolytics reduce osteoblast bone mineralization. This study tested whether antifibrinolytics Amicar and TXA inhibit spine fusion. METHODS Posterolateral L4-L6 fusion was performed in 50 mice, randomized into groups of 10, which received the following treatment before and after surgery: (1) saline; (2) TXA 100 mg/kg; (3) TXA 1000 mg/kg; (4) Amicar 100 mg/kg; and (5) Amicar 1000 mg/kg. High-resolution plane radiography was performed after 5 weeks and micro-CT (computed tomography) was performed at the end of the 12-week study. Radiographs were graded using the Lenke scale. Micro-CT was used to quantify fusion mass bone volume. One-way analysis of variance by ranks with Kruskal-Wallis testing was used to compare the radiographical scores. One-way analysis of variance with least significant difference post hoc testing was used to compare the micro-CT bone volume. RESULTS The average±standard deviation bone volume/total volume (%) measured in the saline, TXA 100 mg/kg, TXA 1000 mg/kg, Amicar 100 mg/kg, and Amicar 1000 mg/kg groups were 10.8±2.3%, 9.7±2.2%, 13.4±3.2%, 15.5±5.2%, and 17.9±3.5%, respectively. There was a significant difference in the Amicar 100 mg/kg (P<0.05) and Amicar 1000 mg/kg (P<0.001) groups compared with the saline group. There was greater bone volume in the Amicar groups compared with the TXA group (P<0.001). There was more bone volume in the TXA 1000 mg/kg group compared with TXA 100 mg/kg (P<0.05) but the bone volume in neither of the TXA groups was different to saline (P=0.49). There were no between-group differences observed using plane radiographical scoring. CONCLUSION Amicar significantly "enhanced" the fusion bone mass in a dose-dependent manner, whereas TXA did not have a significant effect on fusion compared with saline control.These data are in contrast to prior in vitro data that antifibrinolytics inhibit osteoblast bone mineralization. LEVEL OF EVIDENCE N/A.
Collapse
|
30
|
Kawao N, Tamura Y, Okumoto K, Yano M, Okada K, Matsuo O, Kaji H. Tissue-type plasminogen activator deficiency delays bone repair: roles of osteoblastic proliferation and vascular endothelial growth factor. Am J Physiol Endocrinol Metab 2014; 307:E278-88. [PMID: 24918201 DOI: 10.1152/ajpendo.00129.2014] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Further development in research of bone regeneration is necessary to meet the clinical demand for bone reconstruction. Recently, we reported that plasminogen is crucial for bone repair through enhancement of vessel formation. However, the details of the role of tissue-type plasminogen activator (tPA) and urokinase-type plasminogen activator (uPA) in the bone repair process still remain unknown. Herein, we examined the effects of plasminogen activators on bone repair after a femoral bone defect using tPA-deficient (tPA(-/-)) and uPA-deficient (uPA(-/-)) mice. Bone repair of the femur was delayed in tPA(-/-) mice, unlike that in wild-type (tPA(+/+)) mice. Conversely, the bone repair was comparable between wild-type (uPA(+/+)) and uPA(-/-) mice. The number of proliferative osteoblasts was decreased at the site of bone damage in tPA(-/-) mice. Moreover, the proliferation of primary calvarial osteoblasts was reduced in tPA(-/-) mice. Recombinant tPA facilitated the proliferation of mouse osteoblastic MC3T3-E1 cells. The proliferation enhanced by tPA was antagonized by the inhibition of endogenous annexin 2 by siRNA and by the inhibition of extracellular signal-regulated kinase (ERK)1/2 phosphorylation in MC3T3-E1 cells. Vessel formation as well as the levels of vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α) were decreased at the damaged site in tPA(-/-) mice. Our results provide novel evidence that tPA is crucial for bone repair through the facilitation of osteoblast proliferation related to annexin 2 and ERK1/2 as well as enhancement of vessel formation related to VEGF and HIF-1α at the site of bone damage.
Collapse
Affiliation(s)
- Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Yukinori Tamura
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Katsumi Okumoto
- Life Science Research Institute, Kinki University, Osakasayama, Osaka, Japan
| | - Masato Yano
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Osamu Matsuo
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osakasayama, Osaka, Japan; and
| |
Collapse
|
31
|
Khan R, Gupta N, Kumar R, Sharma M, Kumar L, Sharma A. Augmented expression of urokinase plasminogen activator and extracellular matrix proteins associates with multiple myeloma progression. Clin Exp Metastasis 2014; 31:585-93. [PMID: 24807734 DOI: 10.1007/s10585-014-9652-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2013] [Accepted: 03/26/2014] [Indexed: 10/25/2022]
Abstract
Multiple myeloma (MM) represents a B cell malignancy, characterized by a monoclonal proliferation of malignant plasma cells. Interactions between tumor cells and extracellular matrix (ECM) are of importance for tumor invasion and metastasis. Protein levels of urokinase plasminogen activator (uPA) and fibulin 1, nidogen and laminin in plasma and serum respectively and mRNA levels of these molecules in peripheral blood mononuclear cells were determined in 80 subjects by using ELISA and quantitative PCR and data was analyzed with severity of disease. Pearson correlation was determined to observe interrelationship between different molecules. A statistical significant increase for ECM proteins (laminin, nidogen and fibulin 1) and uPA at circulatory level as well as at mRNA level was observed compared to healthy controls. The levels of these molecules in serum might be utilized as a marker of active disease. Significant positive correlation of all ECM proteins with uPA was found and data also correlates with severity of disease. Strong association found between ECM proteins and uPA in this study supports that there might be interplay between these molecules which can be targeted. This study on these molecules may help to gain insight into processes of growth, spread, and clinical behavior of MM.
Collapse
Affiliation(s)
- Rehan Khan
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi, 110029, India
| | | | | | | | | | | |
Collapse
|
32
|
Popa NL, Wergedal JE, Lau KHW, Mohan S, Rundle CH. Urokinase plasminogen activator gene deficiency inhibits fracture cartilage remodeling. J Bone Miner Metab 2014; 32:124-35. [PMID: 23700285 DOI: 10.1007/s00774-013-0475-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2012] [Accepted: 04/18/2013] [Indexed: 01/08/2023]
Abstract
Urokinase plasminogen activator (uPA) regulates a proteolytic cascade of extracellular matrix degradation that functions in tissue development and tissue repair. The development and remodeling of the skeletal extracellular matrix during wound healing suggests that uPA might regulate bone development and repair. To determine whether uPA functions regulate bone development and repair, we examined the basal skeletal phenotype and endochondral bone fracture repair in uPA-deficient mice. The skeletal phenotype of uPA knockout mice was compared with that of control mice under basal conditions by dual-energy X-ray absorptiometry and micro-CT analysis, and during femur fracture repair by micro-CT and histological examination of the fracture callus. No effects of uPA gene deficiency were observed in the basal skeletal phenotype of the whole body or the femur. However, uPA gene deficiency resulted in increased fracture callus cartilage abundance during femur fracture repair at 14 days healing. The increase in cartilage corresponded to reduced tartrate-resistant acid phosphatase (TRAP) staining for osteoclasts in the uPA knockout fracture callus at this time, consistent with impaired osteoclast-mediated remodeling of the fracture cartilage. CD31 staining was reduced in the knockout fracture tissues at this time, suggesting that angiogenesis was also reduced. Osteoclasts also colocalized with CD31 expression in the endothelial cells of the fracture tissues during callus remodeling. These results indicate that uPA promotes remodeling of the fracture cartilage by osteoclasts that are associated with angiogenesis and suggest that uPA promotes angiogenesis and remodeling of the fracture cartilage at this time of bone fracture repair.
Collapse
Affiliation(s)
- Nicoleta L Popa
- Musculoskeletal Disease Center, Research Service (151), Jerry L. Pettis Memorial Veterans Administration Medical Center, 11201 Benton Street, Loma Linda, CA, 92357, USA
| | | | | | | | | |
Collapse
|
33
|
Wyganowska-Świątkowska M, Surdacka A, Skrzypczak-Jankun E, Jankun J. The plasminogen activation system in periodontal tissue (Review). Int J Mol Med 2014; 33:763-8. [PMID: 24535478 DOI: 10.3892/ijmm.2014.1653] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 01/28/2014] [Indexed: 11/05/2022] Open
Abstract
The plasminogen activation system (PAS) plays an essential role in tissue proteolysis in physiological and pathological processes. Periodontitis is a chronic infection associated with increased proteolysis driven by plasminogen activation. In this comprehensive review, we summarise the effects of PAS in wound healing, tissue remodelling, inflammation, bacterial infection, and in the initiation and progression of periodontal disease. Specifically, we discuss the role of plasminogen activators (PAs), including urokinase PA (uPA), tissue-type PA (tPA), PA inhibitor type 1 (PAI-1) and 2 (PAI-2) and activated plasminogen in periodontal tissue, where their concentrations can reach much higher values than those found in other parts of the body. We also discuss whether PA deficiencies can have effects on periodontal tissue. We conclude that in periodontal disease, PAS is unbalanced and equalizing its function can improve the clinical periodontal tissue condition.
Collapse
Affiliation(s)
| | - Anna Surdacka
- Department of Conservative Dentistry and Periodontology, Poznań University of Medical Sciences, Poznań 60-820, Poland
| | - Ewa Skrzypczak-Jankun
- Urology Research Center, Department of Urology, College of Medicine, University of Toledo, Toledo, OH 43614, USA
| | - Jerzy Jankun
- Protein Research Chair, Department of Biochemistry, College of Sciences, King Saud University, Riyadh 11451, Kingdom of Saudi Arabia
| |
Collapse
|
34
|
Graham JR, Chamberland A, Lin Q, Li XJ, Dai D, Zeng W, Ryan MS, Rivera-Bermúdez MA, Flannery CR, Yang Z. Serine protease HTRA1 antagonizes transforming growth factor-β signaling by cleaving its receptors and loss of HTRA1 in vivo enhances bone formation. PLoS One 2013; 8:e74094. [PMID: 24040176 PMCID: PMC3770692 DOI: 10.1371/journal.pone.0074094] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 07/26/2013] [Indexed: 11/19/2022] Open
Abstract
HTRA1 is a member of the High Temperature Requirement (HTRA1) family of serine proteases, which play a role in several biological and pathological processes. In part, HTRA1 regulation occurs by inhibiting the TGF-β signaling pathway, however the mechanism of inhibition has not been fully defined. Previous studies have shown that HTRA1 is expressed in a variety of tissues, including sites of skeletal development. HTRA1 has also been implicated in the process of bone formation, although the precise manner of regulation is still unknown. This study investigated how HTRA1 regulates TGF-β signaling and examined the in vivo effects of the loss of HTRA1. We demonstrated that recombinant HTRA1 was capable of cleaving both type II and type III TGF-β receptors (TβRII and TβRIII) in vitro in a dose-dependent manner, but it did not affect the integrity of TβRI or TGF-β. Overexpression of HTRA1 led to decreased levels of both TβRII and III on the cell surface but had no effect on TβRI. Silencing HTRA1 expression significantly increased TGF-β binding to the cell surface and TGF-β responsiveness within the cell. To examine the role of HTRA1 in vivo, we generated mice with a targeted gene deletion of HTRA1. Embryonic fibroblasts isolated from these mice displayed an increase in TGF-β-induced expression of several genes known to promote bone formation. Importantly, the loss of HTRA1 in the knockout mice resulted in a marked increase in trabecular bone mass. This study has identified a novel regulatory mechanism by which HTRA1 antagonizes TGF-β signaling, and has shown that HTRA1 plays a key role in the regulation of bone formation.
Collapse
Affiliation(s)
- Julie R. Graham
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Angela Chamberland
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Qingcong Lin
- Global BioTherapeutic Technologies, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - X. Jian Li
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - David Dai
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Weilan Zeng
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Mark S. Ryan
- Immunoscience Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Moisés A. Rivera-Bermúdez
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Carl R. Flannery
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
| | - Zhiyong Yang
- Inflammation and Remodeling Research Unit, Pfizer BioTherapeutics Research, Cambridge, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
35
|
Tamura Y, Kawao N, Okada K, Yano M, Okumoto K, Matsuo O, Kaji H. Plasminogen activator inhibitor-1 is involved in streptozotocin-induced bone loss in female mice. Diabetes 2013; 62:3170-9. [PMID: 23715621 PMCID: PMC3749344 DOI: 10.2337/db12-1552] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
In diabetic patients, the risk of fracture is high because of impaired bone formation. However, the details of the mechanisms in the development of diabetic osteoporosis remain unclear. In the current study, we investigated the role of plasminogen activator inhibitor (PAI)-1 in the pathogenesis of type 1 diabetic osteoporosis by using PAI-1-deficient mice. Quantitative computed tomography analysis showed that PAI-1 deficiency protected against streptozotocin-induced bone loss in female mice but not in male mice. PAI-1 deficiency blunted the changes in the levels of Runx2, osterix, and alkaline phosphatase in tibia as well as serum osteocalcin levels suppressed by the diabetic state in female mice only. Furthermore, the osteoclast levels in tibia, suppressed in diabetes, were also blunted by PAI-1 deficiency in female mice. Streptozotocin markedly elevated the levels of PAI-1 mRNA in liver in female mice only. In vitro study demonstrated that treatment with active PAI-1 suppressed the levels of osteogenic genes and mineralization in primary osteoblasts from female mouse calvaria. In conclusion, the current study indicates that PAI-1 is involved in the pathogenesis of type 1 diabetic osteoporosis in females. The expression of PAI-1 in the liver and the sensitivity of bone cells to PAI-1 may be an underlying mechanism.
Collapse
Affiliation(s)
- Yukinori Tamura
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Kiyotaka Okada
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Masato Yano
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Katsumi Okumoto
- Life Science Research Institute, Kinki University, Osaka, Japan
| | - Osamu Matsuo
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | - Hiroshi Kaji
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
- Corresponding author: Hiroshi Kaji,
| |
Collapse
|
36
|
Kawao N, Tamura Y, Okumoto K, Yano M, Okada K, Matsuo O, Kaji H. Plasminogen plays a crucial role in bone repair. J Bone Miner Res 2013; 28:1561-74. [PMID: 23456978 DOI: 10.1002/jbmr.1921] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Revised: 02/02/2013] [Accepted: 02/21/2013] [Indexed: 12/15/2022]
Abstract
The further development in research of bone regeneration is necessary to meet the clinical demand for bone reconstruction. Plasminogen is a critical factor of the tissue fibrinolytic system, which mediates tissue repair in the skin and liver. However, the role of the fibrinolytic system in bone regeneration remains unknown. Herein, we investigated bone repair and ectopic bone formation using plasminogen-deficient (Plg⁻/⁻) mice. Bone repair of the femur is delayed in Plg⁻/⁻ mice, unlike that in the wild-type (Plg⁺/⁺) mice. The deposition of cartilage matrix and osteoblast formation were both decreased in Plg⁻/⁻ mice. Vessel formation, macrophage accumulation, and the levels of vascular endothelial growth factor (VEGF) and transforming growth factor-β (TGF-β) were decreased at the site of bone damage in Plg⁻/⁻ mice. Conversely, heterotopic ossification was not significantly different between Plg⁺/⁺ and Plg⁻/⁻ mice. Moreover, angiogenesis, macrophage accumulation, and the levels of VEGF and TGF-β were comparable between Plg⁺/⁺ and Plg⁻/⁻ mice in heterotopic ossification. Our data provide novel evidence that plasminogen is essential for bone repair. The present study indicates that plasminogen contributes to angiogenesis related to macrophage accumulation, TGF-β, and VEGF, thereby leading to the enhancement of bone repair.
Collapse
Affiliation(s)
- Naoyuki Kawao
- Department of Physiology and Regenerative Medicine, Kinki University Faculty of Medicine, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
37
|
Phospholipases of mineralization competent cells and matrix vesicles: roles in physiological and pathological mineralizations. Int J Mol Sci 2013; 14:5036-129. [PMID: 23455471 PMCID: PMC3634480 DOI: 10.3390/ijms14035036] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Revised: 01/24/2013] [Accepted: 01/25/2013] [Indexed: 02/08/2023] Open
Abstract
The present review aims to systematically and critically analyze the current knowledge on phospholipases and their role in physiological and pathological mineralization undertaken by mineralization competent cells. Cellular lipid metabolism plays an important role in biological mineralization. The physiological mechanisms of mineralization are likely to take place in tissues other than in bones and teeth under specific pathological conditions. For instance, vascular calcification in arteries of patients with renal failure, diabetes mellitus or atherosclerosis recapitulates the mechanisms of bone formation. Osteoporosis—a bone resorbing disease—and rheumatoid arthritis originating from the inflammation in the synovium are also affected by cellular lipid metabolism. The focus is on the lipid metabolism due to the effects of dietary lipids on bone health. These and other phenomena indicate that phospholipases may participate in bone remodelling as evidenced by their expression in smooth muscle cells, in bone forming osteoblasts, chondrocytes and in bone resorbing osteoclasts. Among various enzymes involved, phospholipases A1 or A2, phospholipase C, phospholipase D, autotaxin and sphingomyelinase are engaged in membrane lipid remodelling during early stages of mineralization and cell maturation in mineralization-competent cells. Numerous experimental evidences suggested that phospholipases exert their action at various stages of mineralization by affecting intracellular signaling and cell differentiation. The lipid metabolites—such as arachidonic acid, lysophospholipids, and sphingosine-1-phosphate are involved in cell signaling and inflammation reactions. Phospholipases are also important members of the cellular machinery engaged in matrix vesicle (MV) biogenesis and exocytosis. They may favour mineral formation inside MVs, may catalyse MV membrane breakdown necessary for the release of mineral deposits into extracellular matrix (ECM), or participate in hydrolysis of ECM. The biological functions of phospholipases are discussed from the perspective of animal and cellular knockout models, as well as disease implications, development of potent inhibitors and therapeutic interventions.
Collapse
|
38
|
Kallala R, Barrow J, Graham SM, Kanakaris N, Giannoudis PV. The in vitro and in vivo effects of nicotine on bone, bone cells and fracture repair. Expert Opin Drug Saf 2013; 12:209-33. [PMID: 23410538 DOI: 10.1517/14740338.2013.770471] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION Cigarette smoke has negative effects on bone metabolism and fracture repair. However, no study has reviewed effects of nicotine on bone and fracture repair independent of other constituents of cigarette smoke. The authors review the existing evidence of the effect of nicotine on 'bone' and 'bone cells' and fracture repair, drawing conclusions relevant to clinical practice and future research. AREAS COVERED A literature review was conducted using PRISMA guidelines and PubMed, Cochrane, MEDLINE/OVID, EMBASE, NHS Evidence and Google scholar databases. Articles were included if they specifically investigated the effects of nicotine on 'bone' or fracture repair in animal or human models or in vitro effects on 'bone cells'. A total of 64 papers were included in this review, of which 15 were human in vitro studies and 49 animal studies wherein 9 were in vitro and 40 in vivo. In vivo studies of the effects of nicotine in animals demonstrated widespread effects on bone including osteoneogenesis, osseointegration, steady-state skeletal bone and genes and cytokines relevant to bone cell physiology and bone homeostasis. In these studies, nicotine's effects are predominately negative, inhibiting bone cell metabolism and fracture repair, whereas most in vitro studies reported biphasic responses in all bone cells except osteoclastic cells. EXPERT OPINION The review suggests that nicotine has effects on osteoneogenesis, osseointegration and steady-state skeletal bone in animal in vivo models, as well as effects on all 'bone cells', via several mechanisms in both animal and human cell in vitro studies. The effect of nicotine is dose-dependent, with higher concentrations having predominantly negative effects, whereas at low concentrations a stimulatory effect is seen. Stimulatory effects on certain cells may indicate a possible, limited therapeutic role; advice regarding smoking cessation perioperatively should remain due to the other harmful components of cigarette smoke, but there may be scope for allowing the use of nicotine patches instead of complete abstention. Further research into clinical outcomes is required before the exact response of bone and fracture repair in humans to nicotine is known.
Collapse
Affiliation(s)
- Rami Kallala
- University of Leeds, School of Medicine, Academic Department of Orthopaedic and Trauma Surgery, Leeds General Infirmary, Gt. George Street, LS1 3EX, Leeds, UK.
| | | | | | | | | |
Collapse
|
39
|
Sulniute R, Lindh T, Wilczynska M, Li J, Ny T. Plasmin is essential in preventing periodontitis in mice. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:819-28. [PMID: 21704601 PMCID: PMC3157224 DOI: 10.1016/j.ajpath.2011.05.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2010] [Revised: 04/03/2011] [Accepted: 05/02/2011] [Indexed: 12/18/2022]
Abstract
Periodontitis involves bacterial infection, inflammation of the periodontium, degradation of gum tissue, and alveolar bone resorption, which eventually leads to loss of teeth. To study the role of the broad-spectrum protease plasmin in periodontitis, we examined the oral health of plasminogen (Plg)-deficient mice. In wild-type mice, the periodontium was unaffected at all time points studied; in Plg-deficient mice, periodontitis progressed rapidly, within 20 weeks. Morphological study results of Plg-deficient mice revealed detachment of gingival tissue, resorption of the cementum layer, formation of necrotic tissue, and severe alveolar bone degradation. IHC staining showed massive infiltration of neutrophils in the periodontal tissues. Interestingly, doubly deficient mice, lacking both tissue- and urokinase-type plasminogen activators, developed periodontal disease similar to that in Plg-deficient mice; however, mice lacking only tissue- or urokinase-type plasminogen activator remained healthy. Supplementation by injection of Plg-deficient mice with human plasminogen for 10 days led to necrotic tissue absorption, inflammation subsidence, and full regeneration of gum tissues. Notably, there was also partial regrowth of degraded alveolar bone. Taken together, our results show that plasminogen is essential for the maintenance of a healthy periodontium and plays an important role in combating the spontaneous development of chronic periodontitis. Moreover, reversal to healthy status after supplementation of Plg-deficient mice with plasminogen suggests the possibility of using plasminogen for therapy of periodontal diseases.
Collapse
Affiliation(s)
- Rima Sulniute
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Tomas Lindh
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
- Department of Odontology/Prosthetic Dentistry, Umeå University, Umeå, Sweden
| | | | - Jinan Li
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| | - Tor Ny
- Department of Medical Biochemistry and Biophysics, Umeå University, Umeå, Sweden
| |
Collapse
|
40
|
Kanno Y, Ishisaki A, Kawashita E, Chosa N, Nakajima K, Nishihara T, Toyoshima K, Okada K, Ueshima S, Matsushita K, Matsuo O, Matsuno H. Plasminogen/plasmin modulates bone metabolism by regulating the osteoblast and osteoclast function. J Biol Chem 2011; 286:8952-60. [PMID: 21239499 DOI: 10.1074/jbc.m110.152181] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The contribution of plasminogen (Plg)/plasmin, which have claimed to be the main fibrinolytic regulators in the bone metabolism, remains unclear. This study evaluated how the absence of Plg affects the function of osteoblast (OB) and osteoclast (OC). There was a larger population of pre-OCs in bone marrow-derived cells from the Plg(-/-) mice than the population of that from the WT mice. In addition, the absence of Plg suppressed the expression of osteoprotegerin in OBs. Moreover, an exogenous plasmin clearly induced the osteoprotegerin expression in Plg(-/-) OBs. The osteoclastogenesis of RAW264.7 mouse monocyte/macrophage lineage cells in co-culture with OBs from the Plg(-/-) mice was significantly accelerated in comparison with that in co-culture with OBs from the WT mice. Intriguingly, the accelerated OC differentiation of RAW264.7 cells co-cultured with Plg(-/-) OBs was clearly suppressed by the treatment of an exogenous plasmin. Consequently, Plg(-/-) mice display decreased bone mineral density. These findings could eventually lead to the development of new clinical therapies for bone disease caused by a disorder of the fibrinolytic system.
Collapse
Affiliation(s)
- Yosuke Kanno
- Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's Collage of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Bajaj MS, Ogueli GI, Kumar Y, Vadivel K, Lawson G, Shanker S, Schmidt AE, Bajaj SP. Engineering kunitz domain 1 (KD1) of human tissue factor pathway inhibitor-2 to selectively inhibit fibrinolysis: properties of KD1-L17R variant. J Biol Chem 2010; 286:4329-40. [PMID: 21115497 DOI: 10.1074/jbc.m110.191163] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tissue factor pathway inhibitor-2 (TFPI-2) inhibits factor XIa, plasma kallikrein, and factor VIIa/tissue factor; accordingly, it has been proposed for use as an anticoagulant. Full-length TFPI-2 or its isolated first Kunitz domain (KD1) also inhibits plasmin; therefore, it has been proposed for use as an antifibrinolytic agent. However, the anticoagulant properties of TFPI-2 or KD1 would diminish its antifibrinolytic function. In this study, structure-based investigations and analysis of the serine protease profiles revealed that coagulation enzymes prefer a hydrophobic residue at the P2' position in their substrates/inhibitors, whereas plasmin prefers a positively charged arginine residue at the corresponding position in its substrates/inhibitors. Based upon this observation, we changed the P2' residue Leu-17 in KD1 to Arg (KD1-L17R) and compared its inhibitory properties with wild-type KD1 (KD1-WT). Both WT and KD1-L17R were expressed in Escherichia coli, folded, and purified to homogeneity. N-terminal sequences and mass spectra confirmed proper expression of KD1-WT and KD1-L17R. Compared with KD1-WT, the KD1-L17R did not inhibit factor XIa, plasma kallikrein, or factor VIIa/tissue factor. Furthermore, KD1-L17R inhibited plasmin with ∼6-fold increased affinity and effectively prevented plasma clot fibrinolysis induced by tissue plasminogen activator. Similarly, in a mouse liver laceration bleeding model, KD1-L17R was ∼8-fold more effective than KD1-WT in preventing blood loss. Importantly, in this bleeding model, KD1-L17R was equally or more effective than aprotinin or tranexamic acid, which have been used as antifibrinolytic agents to prevent blood loss during major surgery/trauma. Furthermore, as compared with aprotinin, renal toxicity was not observed with KD1-L17R.
Collapse
Affiliation(s)
- Madhu S Bajaj
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, UCLA, Los Angeles, California 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Ortega N, Wang K, Ferrara N, Werb Z, Vu TH. Complementary interplay between matrix metalloproteinase-9, vascular endothelial growth factor and osteoclast function drives endochondral bone formation. Dis Model Mech 2010; 3:224-35. [PMID: 20142327 DOI: 10.1242/dmm.004226] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Long bone development depends on endochondral bone formation, a complex process requiring exquisite balance between hypertrophic cartilage (HC) formation and its ossification. Dysregulation of this process may result in skeletal dysplasias and heterotopic ossification. Endochondral ossification requires the precise orchestration of HC vascularization, extracellular matrix remodeling, and the recruitment of osteoclasts and osteoblasts. Matrix metalloproteinase-9 (MMP-9), vascular endothelial growth factor (VEGF) and osteoclasts have all been shown to regulate endochondral ossification, but how their function interrelates is not known. We have investigated the functional relationship among these regulators of endochondral ossification, demonstrating that they have complementary but non-overlapping functions. MMP-9, VEGF and osteoclast deficiency all cause impaired growth plate ossification resulting in the accumulation of HC. VEGF mRNA and protein expression are increased at the MMP-9-/- growth plate, and VEGF activity contributes to endochondral ossification since sequestration of VEGF by soluble receptors results in further inhibition of growth plate vascularization and ossification. However, VEGF bioavailability is still limited in MMP-9 deficiency, as exogenous VEGF is able to rescue the MMP-9-/- phenotype, demonstrating that MMP-9 may partially, but not fully, regulate VEGF bioavailability. The organization of the HC extracellular matrix at the MMP-9-/- growth plate is altered, supporting a role for MMP-9 in HC remodeling. Inhibition of VEGF impairs osteoclast recruitment, whereas MMP-9 deficiency leads to an accumulation of osteoclasts at the chondro-osseous junction. Growth plate ossification in osteoclast-deficient mice is impaired in the presence of normal MMP-9 expression, indicating that other osteoclastic functions are also necessary. Our data delineate the complementary interplay between MMP-9, VEGF and osteoclast function that is necessary for normal endochondral bone formation and provide a molecular framework for investigating the molecular defects contributing to disorders of endochondral bone formation.
Collapse
Affiliation(s)
- Nathalie Ortega
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| | | | | | | | | |
Collapse
|
43
|
Agis H, Kandler B, Fischer MB, Watzek G, Gruber R. Activated platelets increase fibrinolysis of mesenchymal progenitor cells. J Orthop Res 2009; 27:972-80. [PMID: 19030175 DOI: 10.1002/jor.20819] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Bone regeneration is initiated by the formation of a blood clot. Activated platelets within this fibrin-rich matrix release signaling molecules that can attract mesenchymal progenitor cells. To gain insight into the cellular mechanism by which activated platelets can support the immigration of mesenchymal progenitors, we have tested the hypothesis that platelet-released signaling molecules increase the capacity of bone marrow stromal cells (BMSC) to activate plasminogen. We report herein that platelet-released supernatants (PRS) elevate total urokinase-type plasminogen activator (uPA) and total plasminogen activator inhibitor-1 (PAI-1) levels in BMSC, as assessed by immunoassay. Quantitative polymerase chain reaction showed an upregulation of uPA, uPA receptor, and PAI-1. Zymography and kinetic analysis based on casein hydrolysis revealed enhanced activity of cell-associated uPA upon exposure of BMSC to PRS. Inhibiting c-Jun N-terminal kinase (JNK) and phosphatidylinositol 3-kinase (PI3K) signaling reduced uPA production and decreased plasminogen activation. Corresponding Western blot analysis showed increased phosphorylation of JNK and AKT in BMSC treated with PRS. These results suggest that activated platelets can enhance the plasminogen activation capacity of mesenchymal progenitors through the stimulation of uPA production, requiring JNK and PI3K/AKT signaling. By this mechanism platelets may contribute to the organization of the blood clot during bone regeneration.
Collapse
Affiliation(s)
- Hermann Agis
- Department of Oral Surgery, Medical University of Vienna, Währingerstrasse 25a, 1090 Vienna, Austria
| | | | | | | | | |
Collapse
|
44
|
Everts V, Daci E, Tigchelaar-Gutter W, Hoeben KA, Torrekens S, Carmeliet G, Beertsen W. Plasminogen activators are involved in the degradation of bone by osteoclasts. Bone 2008; 43:915-20. [PMID: 18691680 DOI: 10.1016/j.bone.2008.07.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2007] [Revised: 06/24/2008] [Accepted: 07/01/2008] [Indexed: 10/21/2022]
Abstract
Osteoclastic bone degradation depends on the activity of several proteolytic enzymes, in particular to those belonging to the classes of cysteine proteinases and matrix metalloproteinases (MMPs). Yet, several findings suggest that the two types of plasminogen activators (PA), the tissue- and urokinase-type PA (tPA and uPA, respectively) are also involved in this process. To investigate the involvement of these enzymes in osteoclast-mediated bone matrix digestion, we analyzed bone explants of mice that were deficient for both tPA and uPA and compared them to wild type mice. The number of osteoclasts as well as their ultrastructural appearance was similar for both genotypes. Next, calvarial and metatarsal bone explants were cultured for 6 or 24 h in the presence of selective inhibitors of cysteine proteinases or MMPs and the effect on osteoclast-mediated bone matrix degradation was assessed. Inhibition of the activity of cysteine proteinases in explants of control mice resulted in massive areas of non-digested demineralized bone matrix adjacent to the ruffled border of osteoclasts, an effect already maximal after 6 h. However, at that time point these demineralized areas were not observed in bone explants from uPA/tPA deficient mice. After prolonged culturing (24 h), a comparable amount of demineralized bone matrix adjacent to actively resorbing osteoclasts was observed in the two genotypes, suggesting that degradation was delayed in uPA/tPA deficient bones. The activity of cysteine proteinases as assessed in bone extracts, proved to be higher in extracts from uPA/tPA(-/-) bones. Immunolocalization of the integrin alpha(v)beta(3) of in vitro generated osteoclasts demonstrated a more diffuse labeling of osteoclasts derived from uPA/tPA(-/-) mice. Taken together, our data indicate that the PAs play a hitherto unrecognized role in osteoclast-mediated bone digestion. The present findings suggest that the PAs are involved in the initial steps of bone degradation, probably by a proper integrin-dependent attachment to bone.
Collapse
Affiliation(s)
- Vincent Everts
- Department of Oral Cell Biology, Academic Centre for Dentistry Amsterdam (ACTA), Universiteit van Amsterdam and Vrije Universiteit, Research Institute MOVE, Van der Boechorststraat 7, 1081 BT Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
45
|
Rundle CH, Wang X, Wergedal JE, Mohan S, Lau KHW. Fracture healing in mice deficient in plasminogen activator inhibitor-1. Calcif Tissue Int 2008; 83:276-84. [PMID: 18820962 DOI: 10.1007/s00223-008-9169-7] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2008] [Accepted: 08/18/2008] [Indexed: 11/26/2022]
Abstract
To evaluate the role of plasminogen activator inhibitor (PAI)-1, a key negative regulator of the plasmin system of extracellular matrix proteases in developmental bone growth and fracture repair, the bone phenotype of male adult PAI-1-deficient mice was determined and femoral fracture healing was compared with that of age- and sex-matched wild-type C57BL/6J control mice. Regarding bone phenotype, the length and size (but not cortical thickness) of the femur of male PAI-1-deficient mice were smaller than those of wild-type controls. Although the total bone mineral content of PAI-1-deficient mice was not significantly different from that of wild-type mice, the total bone area in PAI-1-deficient mice was smaller, leading to an increase in total bone mineral density. With respect to fracture healing, PAI-1-deficient mice developed fracture calluses that were larger and more mineralized than those of wild-type mice but only at 14 days postfracture. These changes were even greater given the smaller size of the normal femur in PAI-1-deficient mice. Surprisingly, the larger fracture callus remodeled rapidly to normal size and mineral content by 21 days postfracture. Examination of fracture histology revealed that these changes were associated with a dramatic increase followed by a rapid remodeling of the fracture callus cartilage. The remodeling of fracture callus cartilage in PAI-1-deficient mice also displayed an abnormal pattern. These findings demonstrate for the first time that PAI-1 (and potentially the plasminogen extracellular matrix protease system) is an important regulator of bone size during developmental growth and plays a regulatory role in the determination of fracture callus size, cartilage formation, and resorption during bone fracture repair.
Collapse
Affiliation(s)
- Charles H Rundle
- Musculoskeletal Disease Center, Jerry L. Pettis Memorial Veterans Administration Medical Center, 11201 Benton Street, Loma Linda, CA 92357, USA
| | | | | | | | | |
Collapse
|
46
|
Madsen CD, Sidenius N. The interaction between urokinase receptor and vitronectin in cell adhesion and signalling. Eur J Cell Biol 2008; 87:617-29. [DOI: 10.1016/j.ejcb.2008.02.003] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 01/31/2008] [Accepted: 02/04/2008] [Indexed: 01/16/2023] Open
|
47
|
Schmidt AE, Sun MF, Ogawa T, Bajaj SP, Gailani D. Functional role of residue 193 (chymotrypsin numbering) in serine proteases: influence of side chain length and beta-branching on the catalytic activity of blood coagulation factor XIa. Biochemistry 2008; 47:1326-35. [PMID: 18186617 DOI: 10.1021/bi701594j] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In serine proteases, Gly193 (chymotrypsin numbering) is conserved with rare exception. Mutants of blood coagulation proteases have been reported with Glu, Ala, Arg or Val substitutions for Gly193. To further understand the role of Gly193 in protease activity, we replaced it with Ala or Val in coagulation factor XIa (FXIa). For comparison to the reported FXIa Glu193 mutant, we prepared FXIa with Asp (short side chain) or Lys (opposite charge) substitutions. Binding of p-aminobenzamidine (pAB) and diisopropylfluorphosphate (DFP) were impaired 1.6-36-fold and 35-478-fold, respectively, indicating distortion of, or altered accessibility to, the S1 and oxyanion-binding sites. Val or Asp substitutions caused the most impairment. Salt bridge formation between the amino terminus of the mature protease moiety at Ile16 and Asp194, essential for catalysis, was impaired 1.4-4-fold. Mutations reduced catalytic efficiency of tripeptide substrate hydrolysis 6-280-fold, with Val or Asp causing the most impairment. Further studies were directed toward macromolecular interactions with the FXIa mutants. kcat for factor IX activation was reduced 8-fold for Ala and 400-1100-fold for other mutants, while binding of the inhibitors antithrombin and amyloid beta-precursor protein Kunitz domain (APPI) was impaired 13-2300-fold and 22-27000-fold, respectively. The data indicate that beta-branching of the side chain of residue 193 is deleterious for interactions with pAB, DFP and amidolytic substrates, situations where no S2'-P2' interactions are involved. When an S2'-P2' interaction is involved (factor IX, antithrombin, APPI), beta-branching and increased side chain length are detrimental. Molecular models indicate that the mutants have impaired S2' binding sites and that beta-branching causes steric conflicts with the FXIa 140-loop, which could perturb the local tertiary structure of the protease domain. In conclusion, enzyme activity is impaired in FXIa when Gly193 is replaced by a non-Gly residue, and residues with side chains that branch at the beta-carbon have the greatest effect on catalysis and binding of substrates.
Collapse
Affiliation(s)
- Amy E Schmidt
- UCLA/Orthopaedic Hospital, Department of Orthopaedic Surgery and Molecular Biology Institute, David Geffen School of Medicine at UCLA, Los Angeles, California 90095, USA
| | | | | | | | | |
Collapse
|
48
|
Nordstrom SM, Carleton SM, Carson WL, Eren M, Phillips CL, Vaughan DE. Transgenic over-expression of plasminogen activator inhibitor-1 results in age-dependent and gender-specific increases in bone strength and mineralization. Bone 2007; 41:995-1004. [PMID: 17888748 PMCID: PMC3933377 DOI: 10.1016/j.bone.2007.08.020] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2007] [Revised: 07/16/2007] [Accepted: 08/04/2007] [Indexed: 11/22/2022]
Abstract
The plasminogen activation system (PAS) and its principal inhibitor, plasminogen activator inhibitor-1 (PAI-1), are recognized modulators of matrix. In addition, the PAS has previously been implicated in the regulation of bone homeostasis. Our objective was to study the influence of active PAI-1 on geometric, biomechanical, and mineral characteristics of bone using transgenic mice that over-express a variant of human PAI-1 that exhibits enhanced functional stability. Femora were isolated from male and female, wildtype (WT) and transgenic (PAI-1.stab) mice at 16 and 32 weeks of age (n=10). Femora were imaged via DEXA for BMD and muCT for cortical mid-slice geometry. Torsional testing was employed for biomechanical properties. Mineral composition was analyzed via instrumental neutron activation analysis. Female femora were further analyzed for trabecular bone histomorphometry (n=11). Whole animal DEXA scans were performed on PAI-1.stab females and additional transgenic lines in which the functional domains of the PAI-1 protein were specifically disrupted. Thirty-two week female PAI-1.stab femora exhibited decreased mid-slice diameters and reduced polar moment of area compared to WT, while maintaining similar cortical bone width. Greater biomechanical strength and stiffness were demonstrated by 32 week PAI-1.stab female femora in addition to a 52% increase in BMD. PAI-1.stab trabecular bone architecture was comparable to WT. Osteoid area was decreased in PAI-1.stab mice while mineral apposition rate increased by 78% over WT. Transgenic mice expressing a reactive-site mutant form of PAI-1 showed an increase in BMD similar to PAI-1.stab, whereas transgenic mice expressing a PAI-1 with reduced affinity for vitronectin were comparable to WT. Over-expression of PAI-1 resulted in increased mineralization and biomechanical properties of mouse femora in an age-dependent and gender-specific manner. Changes in mineral preceded increases in strength/stiffness and deterred normal cross-sectional expansion of cortical bone in females. Trabecular bone was not altered in PAI-1.stab mice whereas MAR increased significantly, further supporting mineral changes as the underlying factor in strength differences. The primary influence of PAI-1 occurred during a period of basal bone remodeling, attributing a role for this system in remodeling as opposed to development. Comparison of transgenic lines indicates that PAI-1's influence on bone is dependent on its ability to bind vitronectin, and not on its proteolytic activity. The impact of PAI-1 on mouse femora supports a regulatory role of the plasminogen activation system in bone homeostasis, potentially elucidating novel targets for the treatment of bone disease.
Collapse
Affiliation(s)
- S M Nordstrom
- Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | | | | | | | | | | |
Collapse
|
49
|
Woodward JKL, Holen I, Coleman RE, Buttle DJ. The roles of proteolytic enzymes in the development of tumour-induced bone disease in breast and prostate cancer. Bone 2007; 41:912-27. [PMID: 17945547 DOI: 10.1016/j.bone.2007.07.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Revised: 07/18/2007] [Accepted: 07/31/2007] [Indexed: 10/22/2022]
Abstract
Tumour-induced bone disease is a common clinical feature of advanced breast and prostate cancer and is associated with considerable morbidity for the affected patients. Our understanding of the molecular mechanisms underlying the development of bone metastases is incomplete, but proteolytic enzymes are implicated in a number of processes involved in both bone metastasis and in normal bone turnover, including matrix degradation, cell migration, angiogenesis, tumour promotion and growth factor activation. Malignant as well as non-malignant cells in the primary and secondary sites express these enzymes, the activity of which may be regulated by soluble factors, cell- or matrix-associated components, as well as a number of cell signalling pathways. A number of secreted and cell surface-associated proteolytic enzymes are implicated in tumour-induced bone disease, including the matrix metalloproteinases, lysosomal cysteine proteinases and plasminogen activators. This review will introduce the role of proteolytic enzymes in normal bone turnover and give an overview of the studies in which their involvement and regulation in the development of bone metastases in breast and prostate cancer has been described. The results from trials involving protease inhibitors in clinical development will also be briefly discussed.
Collapse
Affiliation(s)
- Julia K L Woodward
- Academic Unit of Clinical Oncology, D Floor, School of Medicine and Biomedical Sciences, University of Sheffield, Beech Hill Road, Sheffield, S10 2RX, UK
| | | | | | | |
Collapse
|
50
|
Hecht M, von Metzler I, Sack K, Kaiser M, Sezer O. Interactions of myeloma cells with osteoclasts promote tumour expansion and bone degradation through activation of a complex signalling network and upregulation of cathepsin K, matrix metalloproteinases (MMPs) and urokinase plasminogen activator (uPA). Exp Cell Res 2007; 314:1082-93. [PMID: 18053985 DOI: 10.1016/j.yexcr.2007.10.021] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2007] [Revised: 10/16/2007] [Accepted: 10/26/2007] [Indexed: 01/10/2023]
Abstract
Bone destruction is one of the most debilitating manifestations of multiple myeloma (MM) and results from the interaction of myeloma cells with the bone marrow microenvironment. Within the bone marrow, the disturbed balance between osteoclasts and osteoblasts is important for the development of lytic lesions. However, the mechanisms behind myeloma-mediated bone destruction are not completely understood. In order to address the importance of myeloma cell-osteoclast interactions in MM pathogenesis, we have developed a functional coculture system. We found that myeloma-osteoclast interactions resulted in stimulation of myeloma cell growth and osteoclastic activity through activation of major signalling pathways and upregulation of proteases. Signals from osteoclasts activated the p44/p42 MAPK, STAT3 and PI3K/Akt pathways in myeloma cells. In turn, myeloma cells triggered p38 MAPK and NF-kappaB signalling in osteoclasts. Myeloma-osteoclast interactions stimulated the production of TRAP, cathepsin K, matrix metalloproteinase (MMP)-1, -9, and urokinase plasminogen activator (uPA). Consistent data with myeloma cell lines and primary myeloma cells underlined the biological relevance of these findings. In conclusion, we demonstrated the critical role of myeloma cell-osteoclast interactions in the existing interdependence between tumour expansion and bone disease. The identified molecular events might provide the rationale for novel treatment strategies.
Collapse
Affiliation(s)
- Monica Hecht
- Department of Haematology and Oncology, Charité Universitaetsmedizin Berlin, Charitéplatz 1, Berlin, Germany.
| | | | | | | | | |
Collapse
|