1
|
Akbarzadeh M, Flegel J, Patil S, Shang E, Narayan R, Buchholzer M, Kazemein Jasemi NS, Grigalunas M, Krzyzanowski A, Abegg D, Shuster A, Potowski M, Karatas H, Karageorgis G, Mosaddeghzadeh N, Zischinsky M, Merten C, Golz C, Brieger L, Strohmann C, Antonchick AP, Janning P, Adibekian A, Goody RS, Ahmadian MR, Ziegler S, Waldmann H. The Pseudo-Natural Product Rhonin Targets RHOGDI. Angew Chem Int Ed Engl 2022; 61:e202115193. [PMID: 35170181 PMCID: PMC9313812 DOI: 10.1002/anie.202115193] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Indexed: 11/18/2022]
Abstract
For the discovery of novel chemical matter generally endowed with bioactivity, strategies may be particularly efficient that combine previous insight about biological relevance, e.g., natural product (NP) structure, with methods that enable efficient coverage of chemical space, such as fragment-based design. We describe the de novo combination of different 5-membered NP-derived N-heteroatom fragments to structurally unprecedented "pseudo-natural products" in an efficient complexity-generating and enantioselective one-pot synthesis sequence. The pseudo-NPs inherit characteristic elements of NP structure but occupy areas of chemical space not covered by NP-derived chemotypes, and may have novel biological targets. Investigation of the pseudo-NPs in unbiased phenotypic assays and target identification led to the discovery of the first small-molecule ligand of the RHO GDP-dissociation inhibitor 1 (RHOGDI1), termed Rhonin. Rhonin inhibits the binding of the RHOGDI1 chaperone to GDP-bound RHO GTPases and alters the subcellular localization of RHO GTPases.
Collapse
Affiliation(s)
- Mohammad Akbarzadeh
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
- Institute of Biochemistry and Molecular Biology IIMedical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfUniversitätsstrasse 1, Building 22.03.0540225DüsseldorfGermany
| | - Jana Flegel
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Sumersing Patil
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Erchang Shang
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Rishikesh Narayan
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
- School of Chemical and Materials SciencesIIT Goa, FarmagudiPondaGoa-403401India
| | - Marcel Buchholzer
- Institute of Biochemistry and Molecular Biology IIMedical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfUniversitätsstrasse 1, Building 22.03.0540225DüsseldorfGermany
| | - Neda S. Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology IIMedical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfUniversitätsstrasse 1, Building 22.03.0540225DüsseldorfGermany
| | - Michael Grigalunas
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Adrian Krzyzanowski
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Straße 644221DortmundGermany
| | - Daniel Abegg
- Department of ChemistryThe Scripps Research Institute130 Scripps WayJupiterFL 33458USA
| | - Anton Shuster
- Department of ChemistryThe Scripps Research Institute130 Scripps WayJupiterFL 33458USA
| | - Marco Potowski
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Hacer Karatas
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - George Karageorgis
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Niloufar Mosaddeghzadeh
- Institute of Biochemistry and Molecular Biology IIMedical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfUniversitätsstrasse 1, Building 22.03.0540225DüsseldorfGermany
| | | | - Christian Merten
- Faculty of Chemistry and BiochemistryOrganic Chemistry IIRuhr-University BochumUniversitätsstrasse 15044780BochumGermany
| | - Christopher Golz
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Straße 644221DortmundGermany
| | - Lucas Brieger
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Straße 644221DortmundGermany
| | - Carsten Strohmann
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Straße 644221DortmundGermany
| | - Andrey P. Antonchick
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Petra Janning
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Alexander Adibekian
- Department of ChemistryThe Scripps Research Institute130 Scripps WayJupiterFL 33458USA
| | - Roger S. Goody
- Max Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology IIMedical Faculty and University Hospital DüsseldorfHeinrich Heine University DüsseldorfUniversitätsstrasse 1, Building 22.03.0540225DüsseldorfGermany
| | - Slava Ziegler
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
| | - Herbert Waldmann
- Department of Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Straße 1144227DortmundGermany
- Faculty of Chemistry and Chemical BiologyTechnical University DortmundOtto-Hahn-Straße 644221DortmundGermany
| |
Collapse
|
2
|
Akbarzadeh M, Flegel J, Patil S, Shang E, Narayan R, Buchholzer M, Kazemein Jasemi NS, Grigalunas M, Krzyzanowski A, Abegg D, Shuster A, Potowski M, Karatas H, Karageorgis G, Mosaddeghzadeh N, Zischinsky M, Merten C, Golz C, Brieger L, Strohmann C, Antonchick AP, Janning P, Adibekian A, Goody RS, Ahmadian MR, Ziegler S, Waldmann H. The Pseudo‐Natural Product Rhonin Targets RHOGDI. Angew Chem Int Ed Engl 2022. [DOI: 10.1002/ange.202115193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Mohammad Akbarzadeh
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
- Institute of Biochemistry and Molecular Biology II Medical Faculty and University Hospital Düsseldorf Heinrich Heine University Düsseldorf Universitätsstrasse 1, Building 22.03.05 40225 Düsseldorf Germany
| | - Jana Flegel
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Sumersing Patil
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Erchang Shang
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Rishikesh Narayan
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
- School of Chemical and Materials Sciences IIT Goa, Farmagudi Ponda Goa-403401 India
| | - Marcel Buchholzer
- Institute of Biochemistry and Molecular Biology II Medical Faculty and University Hospital Düsseldorf Heinrich Heine University Düsseldorf Universitätsstrasse 1, Building 22.03.05 40225 Düsseldorf Germany
| | - Neda S. Kazemein Jasemi
- Institute of Biochemistry and Molecular Biology II Medical Faculty and University Hospital Düsseldorf Heinrich Heine University Düsseldorf Universitätsstrasse 1, Building 22.03.05 40225 Düsseldorf Germany
| | - Michael Grigalunas
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Adrian Krzyzanowski
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology Technical University Dortmund Otto-Hahn-Straße 6 44221 Dortmund Germany
| | - Daniel Abegg
- Department of Chemistry The Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Anton Shuster
- Department of Chemistry The Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Marco Potowski
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Hacer Karatas
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - George Karageorgis
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Niloufar Mosaddeghzadeh
- Institute of Biochemistry and Molecular Biology II Medical Faculty and University Hospital Düsseldorf Heinrich Heine University Düsseldorf Universitätsstrasse 1, Building 22.03.05 40225 Düsseldorf Germany
| | | | - Christian Merten
- Faculty of Chemistry and Biochemistry Organic Chemistry II Ruhr-University Bochum Universitätsstrasse 150 44780 Bochum Germany
| | - Christopher Golz
- Faculty of Chemistry and Chemical Biology Technical University Dortmund Otto-Hahn-Straße 6 44221 Dortmund Germany
| | - Lucas Brieger
- Faculty of Chemistry and Chemical Biology Technical University Dortmund Otto-Hahn-Straße 6 44221 Dortmund Germany
| | - Carsten Strohmann
- Faculty of Chemistry and Chemical Biology Technical University Dortmund Otto-Hahn-Straße 6 44221 Dortmund Germany
| | - Andrey P. Antonchick
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Petra Janning
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Alexander Adibekian
- Department of Chemistry The Scripps Research Institute 130 Scripps Way Jupiter FL 33458 USA
| | - Roger S. Goody
- Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Mohammad Reza Ahmadian
- Institute of Biochemistry and Molecular Biology II Medical Faculty and University Hospital Düsseldorf Heinrich Heine University Düsseldorf Universitätsstrasse 1, Building 22.03.05 40225 Düsseldorf Germany
| | - Slava Ziegler
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
| | - Herbert Waldmann
- Department of Chemical Biology Max Planck Institute of Molecular Physiology Otto-Hahn-Straße 11 44227 Dortmund Germany
- Faculty of Chemistry and Chemical Biology Technical University Dortmund Otto-Hahn-Straße 6 44221 Dortmund Germany
| |
Collapse
|
3
|
Granados-Montiel J, Cruz-Lemini M, Rangel-Escareño C, Martinez-Nava G, Landa-Solis C, Gomez-Garcia R, Lopez-Reyes A, Espinosa-Gutierrez A, Ibarra C. SERPINA9 and SERPINB2: Novel Cartilage Lineage Differentiation Markers of Human Mesenchymal Stem Cells with Kartogenin. Cartilage 2021; 12:102-111. [PMID: 30373376 PMCID: PMC7755963 DOI: 10.1177/1947603518809403] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
OBJECTIVE Human mesenchymal stem cells (hMSCs) are a promising source for regenerative medicine, especially mesodermal lineages. Clinical applications require an understanding of the mechanisms for transcriptional control to maintain the desired cell type. The aim of this study was to identify novel markers for differentiation of hMSCs into bone or cartilage with the use of Kartogenin, by RNA analysis using microarray technology, and explore the role of RhoA-Rho associated protein kinase (ROCK) inhibition in these. METHODS Commercial human bone marrow derived primary mesenchymal stem cells were purchased from ATCC. Cells were differentiated in vitro in 2-dimensional cultures using Kartogenin as the main cartilage inducer and bone morphogenetic protein 2 for bone differentiation; cells were cultured with and without ROCK inhibitor Y-27632. After 21 days of culture, whole RNA was extracted and analyzed via Affimetrix microarrays. The most significant hits were validated by quantitative polymerase chain reaction. RESULTS We found a total of 1,757 genes that were either up- or downregulated on differentiation, when compared to P1 hMSC (control) at day 0 of differentiation. Two members of the Serpin superfamily, SERPINA9 and SERPINB2, were significantly upregulated in the cartilage groups, whereas they were unchanged in the bone groups with and without ROCK inhibition. CONCLUSIONS SERPINA9 and SERPINB2 are novel differentiation markers, and molecular regulator candidates for hMSC lineage commitment toward bone and cartilage, providing a new tool for regenerative medicine. Our study highlights the roles of these 2 genes, with significant upregulation of both in cell cultures stimulated with Kartogenin.
Collapse
Affiliation(s)
- Julio Granados-Montiel
- Tissue Engineering, Cell Therapy and Regenerative Medicine Research Unit, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico,Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London, UK
| | - Monica Cruz-Lemini
- Fetal Medicine Mexico Foundation and Fetal Surgery Unit, Children and Women’s Specialty Hospital of Queretaro, Queretaro, Mexico
| | | | - Gabriela Martinez-Nava
- Synovioanalysis Molecular Laboratory, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico
| | - Carlos Landa-Solis
- Tissue Engineering, Cell Therapy and Regenerative Medicine Research Unit, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico
| | - Ricardo Gomez-Garcia
- Tissue Engineering, Cell Therapy and Regenerative Medicine Research Unit, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico
| | - Alberto Lopez-Reyes
- Synovioanalysis Molecular Laboratory, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico
| | - Alejandro Espinosa-Gutierrez
- Hand Surgery and Microsurgery Department, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico
| | - Clemente Ibarra
- Tissue Engineering, Cell Therapy and Regenerative Medicine Research Unit, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Mexico City, Mexico,Clemente Ibarra, Tissue Engineering, Cell Therapy and Regenerative Medicine Unit, Instituto Nacional de Rehabilitación “Luis Guillermo Ibarra Ibarra”, Calzada Mexico-Xochimilco No. 289, Col. Arenal de Guadalupe, Tlalpan, Mexico City 14389, Mexico.
| |
Collapse
|
4
|
Steering cell behavior through mechanobiology in 3D: A regenerative medicine perspective. Biomaterials 2020; 268:120572. [PMID: 33285439 DOI: 10.1016/j.biomaterials.2020.120572] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 09/04/2020] [Accepted: 11/21/2020] [Indexed: 12/14/2022]
Abstract
Mechanobiology, translating mechanical signals into biological ones, greatly affects cellular behavior. Steering cellular behavior for cell-based regenerative medicine approaches requires a thorough understanding of the orchestrating molecular mechanisms, among which mechanotransducive ones are being more and more elucidated. Because of their wide use and highly mechanotransduction dependent differentiation, this review focuses on mesenchymal stromal cells (MSCs), while also briefly relating the discussed results to other cell types. While the mechanotransduction pathways are relatively well-studied in 2D, much remains unknown of the role and regulation of these pathways in 3D. Ultimately, cells need to be cultured in a 3D environment to create functional de novo tissue. In this review, we explore the literature on the roles of different material properties on cellular behavior and mechanobiology in 2D and 3D. For example, while stiffness plays a dominant role in 2D MSCs differentiation, it seems to be of subordinate importance in 3D MSCs differentiation, where matrix remodeling seems to be key. Also, the role and regulation of some of the main mechanotransduction players are discussed, focusing on MSCs. We have only just begun to fundamentally understand MSCs and other stem cells behavior in 3D and more fundamental research is required to advance biomaterials able to replicate the stem cell niche and control cell activity. This better understanding will contribute to smarter tissue engineering scaffold design and the advancement of regenerative medicine.
Collapse
|
5
|
Rastogi YR, Saini AK, Thakur VK, Saini RV. New Insights into Molecular Links Between Microbiota and Gastrointestinal Cancers: A Literature Review. Int J Mol Sci 2020; 21:E3212. [PMID: 32370077 PMCID: PMC7246717 DOI: 10.3390/ijms21093212] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Revised: 04/27/2020] [Accepted: 04/28/2020] [Indexed: 12/20/2022] Open
Abstract
Despite decades of exhaustive research on cancer, questions about cancer initiation, development, recurrence, and metastasis have still not been completely answered. One of the reasons is the plethora of factors acting simultaneously in a tumour microenvironment, of which not all have garnered attention. One such factor that has long remained understudied and has only recently received due attention is the host microbiota. Our sheer-sized microbiota exists in a state of symbiosis with the body and exerts significant impact on our body's physiology, ranging from immune-system development and regulation to neurological and cognitive development. The presence of our microbiota is integral to our development, but a change in its composition (microbiota dysbiosis) can often lead to adverse effects, increasing the propensity of serious diseases like cancers. In the present review, we discuss environmental and genetic factors that cause changes in microbiota composition, disposing of the host towards cancer, and the molecular mechanisms (such as β-catenin signalling) and biochemical pathways (like the generation of oncogenic metabolites like N-nitrosamines and hydrogen sulphide) that the microbiota uses to initiate or accelerate cancers, with emphasis on gastrointestinal cancers. Moreover, we discuss how microbiota can adversely influence the success of colorectal-cancer chemotherapy, and its role in tumour metastasis. We also attempted to resolve conflicting results obtained for the butyrate effect on tumour suppression in the colon, often referred to as the 'butyrate paradox'. In addition, we suggest the development of microbiota-based biomarkers for early cancer diagnosis, and a few target molecules of which the inhibition can increase the overall chances of cancer cure.
Collapse
Affiliation(s)
- Yash Raj Rastogi
- School of Bioengineering and Food Technology, Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh 173229, India;
| | - Adesh K. Saini
- Faculty of Sciences, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh 173229, India;
| | - Vijay Kumar Thakur
- Biorefining and Advanced Materials Research Centre, Scotland’s Rural College (SRUC), Kings Buildings, Edinburgh, EH9 3JG, UK
| | - Reena V. Saini
- School of Biotechnology, Faculty of Applied Sciences and Biotechnology, Shoolini University of Biotechnology and Management Sciences, Solan, Himachal Pradesh 173229, India
| |
Collapse
|
6
|
Banu A, Lax AJ, Grigoriadis AE. In Vivo Targets of Pasteurella Multocida Toxin. Int J Mol Sci 2020; 21:ijms21082739. [PMID: 32326543 PMCID: PMC7215291 DOI: 10.3390/ijms21082739] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 01/03/2023] Open
Abstract
Many Pasteurella multocida strains are carried as commensals, while some cause disease in animals and humans. Some type D strains cause atrophic rhinitis in pigs, where the causative agent is known to be the Pasteurella multocida toxin (PMT). PMT activates three families of G-proteins—Gq/11, G12/13, and Gi/o—leading to cellular mitogenesis and other sequelae. The effects of PMT on whole animals in vivo have been investigated previously, but only at the level of organ-specific pathogenesis. We report here the first study to screen all the organs targeted by the toxin by using the QE antibody that recognizes only PMT-modified G-proteins. Under our experimental conditions, short-term treatment of PMT is shown to have multiple in vivo targets, demonstrating G-alpha protein modification, stimulation of proliferation markers and expression of active β-catenin in a tissue- and cell-specific manner. This highlights the usefulness of PMT as an important tool for dissecting the specific roles of different G-alpha proteins in vivo.
Collapse
Affiliation(s)
- Arshiya Banu
- Department of Microbiology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Alistair J. Lax
- Department of Microbiology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
| | - Agamemnon E. Grigoriadis
- Centre for Craniofacial and Regenerative Biology, King’s College London, Guy’s Hospital, London SE1 9RT, UK
- Correspondence: ; Tel.: +44-(0)20-7188-1807
| |
Collapse
|
7
|
Wan Hasan WN, Chin KY, Abd Ghafar N, Soelaiman IN. Annatto-Derived Tocotrienol Promotes Mineralization of MC3T3-E1 Cells by Enhancing BMP-2 Protein Expression via Inhibiting RhoA Activation and HMG-CoA Reductase Gene Expression. Drug Des Devel Ther 2020; 14:969-976. [PMID: 32184566 PMCID: PMC7060796 DOI: 10.2147/dddt.s224941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Accepted: 02/14/2020] [Indexed: 12/14/2022] Open
Abstract
PURPOSE Annatto-derived tocotrienol (AnTT) has been shown to improve bone formation in animal models of osteoporosis and promote differentiation of pre-osteoblastic cells. However, the mechanism of action of AnTT in achieving these effects is unclear. This study aims to investigate the mechanism of action of AnTT on MC3T3-E1 pre-osteoblasts via the mevalonate pathway. METHODS Murine pre-osteoblastic cells, MC3T3-E1, were cultured with the density of 1 × 104 cells/mL and treated with 4 concentrations of AnTT (0.001-1 µg/mL). Expression of HMG-CoA reductase (HMGR) gene was carried out using qPCR after treatment with AnTT for 21 days. RhoA activation and bone morphogenetic protein-2 (BMP-2) were measured using immunoassay after 9 and 15 days of AnTT treatment. Lovastatin was used as the positive control. Mineralized nodules were detected using Von Kossa staining after 21 days of AnTT treatment. RESULTS The results showed that HMGR was up-regulated in the lovastatin group on day 9 and 21 compared to the control. Lovastatin also inhibited RhoA activation (day 9 and 15) and increased BMP-2 protein (day 15). On the other hand, AnTT at 0.001 μg/mL (day 3) and 0.1 μg/mL (day 21) significantly down-regulated HMGR gene expression compared to the control. On day 21, HMGR gene expression was significantly reduced in all groups compared to day 15. AnTT at 0.1 μg/mL significantly decreased RhoA activation on day 9 compared to the control. AnTT at 1 μg/mL significantly increased BMP-2 protein on day 15 compared to the control (P<0.05). Mineralized calcium nodules were more abundant in AnTT treated groups compared to the control on day 21. CONCLUSION AnTT suppresses the mevalonate pathway by downregulating HMGR gene expression and inhibiting RhoA activation, leading to increased BMP-2 protein in MC3T3-E1 cells. This explains the stimulating effects of AnTT on osteoblast mineralization.
Collapse
Affiliation(s)
- Wan Nuraini Wan Hasan
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, UKM Medical Centre (UKMMC), Kuala Lumpur56000, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, UKM Medical Centre (UKMMC), Kuala Lumpur56000, Malaysia
| | - Norzana Abd Ghafar
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, UKM Medical Centre (UKMMC), Kuala Lumpur56000, Malaysia
| | - Ima Nirwana Soelaiman
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, UKM Medical Centre (UKMMC), Kuala Lumpur56000, Malaysia
| |
Collapse
|
8
|
Banu A, Liu KJ, Lax AJ, Grigoriadis AE. G-Alpha Subunit Abundance and Activity Differentially Regulate β-Catenin Signaling. Mol Cell Biol 2019; 39:MCB.00422-18. [PMID: 30559307 PMCID: PMC6379582 DOI: 10.1128/mcb.00422-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 11/27/2018] [Indexed: 11/29/2022] Open
Abstract
Heterotrimeric G proteins are signal transduction proteins involved in regulating numerous signaling events. In particular, previous studies have demonstrated a role for G-proteins in regulating β-catenin signaling. However, the link between G-proteins and β-catenin signaling is controversial and appears to depend on G-protein specificity. We describe a detailed analysis of a link between specific G-alpha subunits and β-catenin using G-alpha subunit genetic knockout and knockdown approaches. The Pasteurella multocida toxin was utilized as a unique tool to activate G-proteins, with LiCl treatment serving as a β-catenin signaling agonist. The results show that Pasteurella multocida toxin (PMT) significantly enhanced LiCl-induced active β-catenin levels in HEK293T cells and mouse embryo fibroblasts. Evaluation of the effect of specific G-alpha proteins on the regulation of β-catenin showed that Gq/11 and G12/13 knockout cells had significantly higher levels of active and total β-catenin than wild-type cells. The stimulation of active β-catenin by PMT and LiCl was lost upon both constitutive and transient knockdown of G12 and G13 but not Gq Based on our results, we conclude that endogenous G-alpha proteins are negative regulators of active β-catenin; however, PMT-activated G-alpha subunits positively regulate LiCl-induced β-catenin expression in a G12/13-dependent manner. Hence, G-alpha subunit regulation of β-catenin is context dependent.
Collapse
Affiliation(s)
- Arshiya Banu
- Department of Microbiology, King's College London, Guy's Hospital, London, United Kingdom
| | - Karen J Liu
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| | - Alistair J Lax
- Department of Microbiology, King's College London, Guy's Hospital, London, United Kingdom
| | - Agamemnon E Grigoriadis
- Centre for Craniofacial and Regenerative Biology, King's College London, Guy's Hospital, London, United Kingdom
| |
Collapse
|
9
|
Zhao J, Wang S, Lu S, Bao X, Sun J, Yang X. An Enzyme Cascade-Triggered Fluorogenic and Chromogenic Reaction Applied in Enzyme Activity Assay and Immunoassay. Anal Chem 2018; 90:7754-7760. [DOI: 10.1021/acs.analchem.8b01845] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jiahui Zhao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Shasha Lu
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
- University of Science and Technology of China, Hefei, Anhui 230026, China
| | - Xingfu Bao
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Jian Sun
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| | - Xiurong Yang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, China
| |
Collapse
|
10
|
Toda H, Yamamoto M, Uyama H, Tabata Y. Effect of hydrogel elasticity and ephrinB2-immobilized manner on Runx2 expression of human mesenchymal stem cells. Acta Biomater 2017; 58:312-322. [PMID: 28300720 DOI: 10.1016/j.actbio.2017.03.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 02/20/2017] [Accepted: 03/10/2017] [Indexed: 12/22/2022]
Abstract
The objective of this study is to design the manner of ephrinB2 immobilized onto polyacrylamide (PAAm) hydrogels with varied elasticity and evaluate the effect of hydrogels elasticity and the immobilized manner of ephrinB2 on the Runx2 expression of human mesenchymal stem cells (hMSC). The PAAm hydrogels were prepared by the radical polymerization of acrylamide (AAm), and N,N'-methylenebisacrylamide (BIS). By changing the BIS concentration, the elasticity of PAAm hydrogels changed from 1 to 70kPa. For the bio-specific immobilization of ephrinB2, a chimeric protein of ephrinB2 and Fc domain was immobilized onto protein A-conjugated PAAm hydrogels by making use of the bio-specific interaction between the Fc domain and protein A. When hMSC were cultured on the ephrinB2-immobilized PAAm hydrogels with varied elasticity, the morphology of hMSC was of cuboidal shape on the PAAm hydrogels immobilized with ephrinB2 compared with non-conjugated ones, irrespective of the hydrogels elasticity. The bio-specific immobilization of ephrinB2 enhanced the level of Runx2 expression. The expression level was significantly high for the hydrogels of 3.6 and 5.9kPa elasticity with bio-specific immobilization of ephrinB2 compared with other hydrogels with the same elasticity. The hydrogels showed a significantly down-regulated RhoA activity. It is concluded that the Runx2 expression of hMSC is synergistically influenced by the hydrogels elasticity and their immobilized manner of ephrinB2 immobilized. STATEMENT OF SIGNIFICANCE Differentiation fate of mesenchymal stem cells (MSC) is modified by biochemical and biophysical factors, such as elasticity and signal proteins. However, there are few experiments about combinations of them. In this study, to evaluate the synergistic effect of them on cell properties of MSC, we established to design the manner of Eph signal ligand, ephrinB2, immobilized onto polyacrylamide hydrogels with varied elasticity. The gene expression level of an osteogenic maker, Runx2, was enhanced by the immobilized manner, and significantly enhanced for the hydrogels of around 4kPa elasticity with bio-specific immobilization of ephrinB2. This is the novel report describing to demonstrate that the Runx2 expression of MSC is synergistically influenced by the hydrogels elasticity and their manner of ephrinB2 immobilized.
Collapse
|
11
|
Namazi H, Torabi S. Novel Use of Botulinum Toxin to Ameliorate Arthrofibrosis: An Experimental Study in Rabbits. Toxicol Pathol 2017; 35:715-8. [PMID: 17763285 DOI: 10.1080/01926230701481923] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
This study aimed to investigate the effects of intra-articular botulinum toxin in preventing arthrofibrosis. Arthrofibrosis was induced in both stifle joints of 20 rabbits by transecting the anterior cruciate ligament under intramuscular anesthesia with ketamine and xylazine. Intra-articular toxin at a dose of 0.6 ml (50 unit) and physiologic saline solution (0.6 ml) were injected into the right and left stifle joints, respectively, 3 times with a 1-week interval between each injection. The rabbits were euthanized in the 12th week via high dose anesthesia to remove the stifle joint. The severity of adhesions was assessed, applying a universal scoring system. Also the stifle joints were histologically evaluated for fibrosis. With regards to severity of adhesion a significant reduction in the adhesion score was observed in the toxin-treated group in comparison to untreated controls with mean ± SE values of 0.2 ± 0.1 and 2.4 ± 0.2, respectively ( p < 0.01). The histological evaluation showed no significant fibroblast in the toxin-treated group versus dense fibers with mature fibroblasts in the control group. Our results suggest that botulinum toxin demonstrated efficacy in preventing adhesion after knee surgery and all the parameters monitored showed consistent statistically significant improvement.
Collapse
Affiliation(s)
- Hamid Namazi
- Shiraz University of Medical Sciences, Shiraz, Iran.
| | | |
Collapse
|
12
|
Higashi K, Matsuzaki E, Hashimoto Y, Takahashi-Yanaga F, Takano A, Anan H, Hirata M, Nishimura F. Sphingosine-1-phosphate/S1PR2-mediated signaling triggers Smad1/5/8 phosphorylation and thereby induces Runx2 expression in osteoblasts. Bone 2016; 93:1-11. [PMID: 27612439 DOI: 10.1016/j.bone.2016.09.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Revised: 08/31/2016] [Accepted: 09/05/2016] [Indexed: 12/11/2022]
Abstract
Sphingosine-1-phosphate (S1P) is a signaling sphingolipid that also plays crucial roles in bone regeneration. Recently, we reported that the S1P receptors S1PR1 and S1PR2 were mainly expressed in osteoblast-like cells, and that the S1P/S1PR1 signaling pathway up-regulated osteoprotegerin and osteoblast differentiation. However, the involvement of S1P/S1PR2 signaling in osteoblast differentiation is not well understood. Here we investigate the role of S1P/S1PR2-mediated signaling in osteoblast differentiation and clarify the underlying signaling mechanisms. We found that an S1P/S1PR2/Gi-independent signaling pathway activated RhoA activity, leading to phosphorylation of Smad1/5/8 in mouse osteoblast-like MC3T3-E1 cells and primary osteoblasts. Furthermore, this signaling pathway promoted nuclear translocation of Smad4, and increased the amount of Smad6/7 protein in the nucleus. S1P also up-regulated runt-related transcription factor 2 (Runx2) expression through S1PR2/RhoA/ROCK/Smad1/5/8 signaling. Moreover, we found that S1P partially triggered S1PR2/RhoA/ROCK pathway leading to bone formation in vivo. These findings suggest that S1P induces RhoA activity, leading to the phosphorylation of Smad1/5/8, thereby promoting Runx2 expression and differentiation in osteoblasts. Our findings describe novel molecular mechanisms in S1P/S1PR2-mediated osteoblast differentiation that could aid future studies of bone regeneration.
Collapse
Affiliation(s)
- Katsumasa Higashi
- Periodontal Section, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Etsuko Matsuzaki
- Periodontal Section, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan; Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan.
| | - Yoko Hashimoto
- Periodontal Section, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Fumi Takahashi-Yanaga
- Department of Clinical Pharmacology, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Aiko Takano
- Periodontal Section, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| | - Hisashi Anan
- Section of Operative Dentistry and Endodontology, Department of Odontology, Fukuoka Dental College, Fukuoka, Japan
| | - Masato Hirata
- Laboratory of Molecular and Cellular Biochemistry, Faculty of Dental Science, Kyushu University, Fukuoka, Japan.
| | - Fusanori Nishimura
- Periodontal Section, Division of Oral Rehabilitation, Faculty of Dental Science, Kyushu University, Fukuoka, Japan
| |
Collapse
|
13
|
Elsafadi M, Manikandan M, Dawud RA, Alajez NM, Hamam R, Alfayez M, Kassem M, Aldahmash A, Mahmood A. Transgelin is a TGFβ-inducible gene that regulates osteoblastic and adipogenic differentiation of human skeletal stem cells through actin cytoskeleston organization. Cell Death Dis 2016; 7:e2321. [PMID: 27490926 PMCID: PMC5108308 DOI: 10.1038/cddis.2016.196] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 04/08/2016] [Accepted: 05/30/2016] [Indexed: 12/11/2022]
Abstract
Regenerative medicine is a novel approach for treating conditions in which enhanced bone regeneration is required. We identified transgelin (TAGLN), a transforming growth factor beta (TGFβ)-inducible gene, as an upregulated gene during in vitro osteoblastic and adipocytic differentiation of human bone marrow-derived stromal (skeletal) stem cells (hMSC). siRNA-mediated gene silencing of TAGLN impaired lineage differentiation into osteoblasts and adipocytes but enhanced cell proliferation. Additional functional studies revealed that TAGLN deficiency impaired hMSC cell motility and in vitro transwell cell migration. On the other hand, TAGLN overexpression reduced hMSC cell proliferation, but enhanced cell migration, osteoblastic and adipocytic differentiation, and in vivo bone formation. In addition, deficiency or overexpression of TAGLN in hMSC was associated with significant changes in cellular and nuclear morphology and cytoplasmic organelle composition as demonstrated by high content imaging and transmission electron microscopy that revealed pronounced alterations in the distribution of the actin filament and changes in cytoskeletal organization. Molecular signature of TAGLN-deficient hMSC showed that several genes and genetic pathways associated with cell differentiation, including regulation of actin cytoskeleton and focal adhesion pathways, were downregulated. Our data demonstrate that TAGLN has a role in generating committed progenitor cells from undifferentiated hMSC by regulating cytoskeleton organization. Targeting TAGLN is a plausible approach to enrich for committed hMSC cells needed for regenerative medicine application.
Collapse
Affiliation(s)
- M Elsafadi
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia.,KMEB, Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark
| | - M Manikandan
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - R A Dawud
- Berlin-Brandenburg Center for Regenerative Therapies, Charité-Universitätsmedizin Berlin, Berlin, Germany.,Department of Comparative Medicine, King Faisal Specialist Hospital and Research Centre, Riyadh, Kingdom of Saudi Arabia
| | - N M Alajez
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - R Hamam
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - M Alfayez
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - M Kassem
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia.,KMEB, Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark
| | - A Aldahmash
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia.,KMEB, Department of Endocrinology, University Hospital of Odense and University of Southern Denmark, Odense, Denmark.,Prince Naif Health Research Center, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| | - A Mahmood
- Stem Cells Unit, Department of Anatomy, College of Medicine, King Saud University, Riyadh 11461, Kingdom of Saudi Arabia
| |
Collapse
|
14
|
Wu T, Song HX, Dong Y, Ye Y, Li JH. Intra-articular injections of botulinum toxin a for refractory joint pain: a systematic review and meta-analysis. Clin Rehabil 2016; 31:435-443. [PMID: 27094033 DOI: 10.1177/0269215516644951] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Tao Wu
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hang Zhou, PR China
- Tao Wu and Hai-xin Song contributed equally to this work
| | - Hai-xin Song
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hang Zhou, PR China
- Tao Wu and Hai-xin Song contributed equally to this work
| | - Yan Dong
- Department of Rehabilitation Medicine, Hangzhou Hospital of Zhejiang CAPF, Hang Zhou, PR China
| | - Ye Ye
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hang Zhou, PR China
| | - Jian-hua Li
- Department of Rehabilitation Medicine, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hang Zhou, PR China
| |
Collapse
|
15
|
Staines KA, Prideaux M, Allen S, Buttle DJ, Pitsillides AA, Farquharson C. E11/Podoplanin Protein Stabilization Through Inhibition of the Proteasome Promotes Osteocyte Differentiation in Murine in Vitro Models. J Cell Physiol 2015; 231:1392-404. [PMID: 26639105 PMCID: PMC4832367 DOI: 10.1002/jcp.25282] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Accepted: 12/04/2015] [Indexed: 11/08/2022]
Abstract
The transmembrane glycoprotein E11 is considered critical in early osteoblast-osteocyte transitions (osteocytogenesis), however its function and regulatory mechanisms are still unknown. Using the late osteoblast MLO-A5 cell line we reveal increased E11 protein/mRNA expression (P < 0.001) concomitant with extensive osteocyte dendrite formation and matrix mineralization (P < 0.001). Transfection with E11 significantly increased mRNA levels (P < 0.001), but immunoblotting failed to detect any correlative increases in E11 protein levels, suggestive of post-translational degradation. We found that exogenous treatment of MLO-A5 and osteocytic IDG-SW3 cells with 10 μM ALLN (calpain and proteasome inhibitor) stabilized E11 protein levels and induced a profound increase in osteocytic dendrite formation (P < 0.001). Treatment with other calpain inhibitors failed to promote similar osteocytogenic changes, suggesting that these effects of ALLN rely upon its proteasome inhibitor actions. Accordingly we found that proteasome-selective inhibitors (MG132/lactacystin/ Bortezomib/Withaferin-A) produced similar dose-dependent increases in E11 protein levels in MLO-A5 and primary osteoblast cells. This proteasomal targeting was confirmed by immunoprecipitation of ubiquitinylated proteins, which included E11, and by increased levels of ubiquitinylated E11 protein upon addition of the proteasome inhibitors MG132/Bortezomib. Activation of RhoA, the small GTPase, was found to be increased concomitant with the peak in E11 levels and its downstream signaling was also observed to promote MLO-A5 cell dendrite formation. Our data indicate that a mechanism reliant upon blockade of proteasome-mediated E11 destabilization contributes to osteocytogenesis and that this may involve downstream targeting of RhoA. This work adds to our mechanistic understanding of the factors regulating bone homeostasis, which may lead to future therapeutic approaches.
Collapse
Affiliation(s)
- Katherine A Staines
- Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| | - Matt Prideaux
- The University of Adelaide, North Terrace, Adelaide, Australia
| | - Steve Allen
- Royal Veterinary College, Royal College Street, London, United Kingdom
| | - David J Buttle
- Department of Infection and Immunity, The University of Sheffield Medical School, Beech Hill Road, Sheffield, United Kingdom
| | | | - Colin Farquharson
- Roslin Institute and R(D)SVS, The University of Edinburgh, Easter Bush, Midlothian, United Kingdom
| |
Collapse
|
16
|
Tai IC, Wang YH, Chen CH, Chuang SC, Chang JK, Ho ML. Simvastatin enhances Rho/actin/cell rigidity pathway contributing to mesenchymal stem cells' osteogenic differentiation. Int J Nanomedicine 2015; 10:5881-94. [PMID: 26451103 PMCID: PMC4590348 DOI: 10.2147/ijn.s84273] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Recent studies have indicated that statins induce osteogenic differentiation both in vitro and in vivo. The molecular mechanism of statin-stimulated osteogenesis is unknown. Activation of RhoA signaling increases cytoskeletal tension, which plays a crucial role in the osteogenic differentiation of mesenchymal stem cells. We thus hypothesized that RhoA signaling is involved in simvastatin-induced osteogenesis in bone marrow mesenchymal stem cells. We found that although treatment with simvastatin shifts localization of RhoA protein from the membrane to the cytosol, the treatment still activates RhoA dose-dependently because it reduces the association with RhoGDIα. Simvastatin also increased the expression of osteogenic proteins, density of actin filament, the number of focal adhesions, and cellular tension. Furthermore, disrupting actin cytoskeleton or decreasing cell rigidity by using chemical agents reduced simvastatin-induced osteogenic differentiation. In vivo study also confirms that density of actin filament is increased in simvastatin-induced ectopic bone formation. Our study is the first to demonstrate that maintaining intact actin cytoskeletons and enhancing cell rigidity are crucial in simvastatin-induced osteogenesis. The results suggested that simvastatin, which is an osteoinductive factor and acts by increasing actin filament organization and cell rigidity combined with osteoconductive biomaterials, may benefit stem-cell-based bone regeneration.
Collapse
Affiliation(s)
- I-Chun Tai
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yao-Hsien Wang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chung-Hwan Chen
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
| | - Shu-Chun Chuang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Je-Ken Chang
- Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Orthopedics, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan ; Department of Orthopedics, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung, Taiwan
| | - Mei-Ling Ho
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Physiology, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Orthopaedic Research Center, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan ; Department of Marine Biotechnology and Resources, National Sun Yat-sen UniVersity, Kaohsiung, Taiwan
| |
Collapse
|
17
|
Chen L, Shi K, Frary CE, Ditzel N, Hu H, Qiu W, Kassem M. Inhibiting actin depolymerization enhances osteoblast differentiation and bone formation in human stromal stem cells. Stem Cell Res 2015. [PMID: 26209815 DOI: 10.1016/j.scr.2015.06.009] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Remodeling of the actin cytoskeleton through actin dynamics is involved in a number of biological processes, but its role in human stromal (skeletal) stem cells (hMSCs) differentiation is poorly understood. In the present study, we demonstrated that stabilizing actin filaments by inhibiting gene expression of the two main actin depolymerizing factors (ADFs): Cofilin 1 (CFL1) and Destrin (DSTN) in hMSCs, enhanced cell viability and differentiation into osteoblastic cells (OB) in vitro, as well as heterotopic bone formation in vivo. Similarly, treating hMSC with Phalloidin, which is known to stabilize polymerized actin filaments, increased hMSCs viability and OB differentiation. Conversely, Cytocholasin D, an inhibitor of actin polymerization, reduced cell viability and inhibited OB differentiation of hMSC. At a molecular level, preventing Cofilin phosphorylation through inhibition of LIM domain kinase 1 (LIMK1) decreased cell viability and impaired OB differentiation of hMSCs. Moreover, depolymerizing actin reduced FAK, p38 and JNK activation during OB differentiation of hMSCs, while polymerizing actin enhanced these signaling pathways. Our results demonstrate that the actin dynamic reassembly and Cofilin phosphorylation loop is involved in the control of hMSC proliferation and osteoblasts differentiation.
Collapse
Affiliation(s)
- Li Chen
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark.
| | - Kaikai Shi
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark
| | - Charles Edward Frary
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark
| | - Nicholas Ditzel
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark
| | - Huimin Hu
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark; Department of Spine Surgery, Honghui Hospital, Xi'an JiaoTong University College of Medicine, 710054 Xi'an, China
| | - Weimin Qiu
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark
| | - Moustapha Kassem
- Molecular Endocrinology Laboratory (KMEB), Odense University Hospital, University of South Denmark, DK-5000 Odense C, Denmark; Danish Stem Cell Center (DanStem), Panum Institute, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
18
|
Jafari A, Siersbaek MS, Chen L, Qanie D, Zaher W, Abdallah BM, Kassem M. Pharmacological Inhibition of Protein Kinase G1 Enhances Bone Formation by Human Skeletal Stem Cells Through Activation of RhoA-Akt Signaling. Stem Cells 2015; 33:2219-31. [DOI: 10.1002/stem.2013] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 02/13/2015] [Indexed: 11/05/2022]
Affiliation(s)
- Abbas Jafari
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
- Danish Stem Cell Center (DanStem); Institute of Cellular and Molecular Medicine, University of Copenhagen; Copenhagen Denmark
| | - Majken S. Siersbaek
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
- Danish Stem Cell Center (DanStem); Institute of Cellular and Molecular Medicine, University of Copenhagen; Copenhagen Denmark
| | - Li Chen
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
| | - Diyako Qanie
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
| | - Walid Zaher
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
| | - Basem M. Abdallah
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
| | - Moustapha Kassem
- Department of Endocrinology and Metabolism, Endocrine Research Laboratory (KMEB); Odense University Hospital & University of Southern Denmark; Odense Denmark
- Danish Stem Cell Center (DanStem); Institute of Cellular and Molecular Medicine, University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
19
|
Pilton JL, Ley CJ, Voss K, Krockenberger MB, Barrs VR, Beatty JA. Atypical, abscessated nasopharyngeal polyp associated with expansion and lysis of the tympanic bulla. J Feline Med Surg 2014; 16:699-702. [PMID: 24319059 PMCID: PMC11164163 DOI: 10.1177/1098612x13514421] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
A 5-year-old, male neutered domestic shorthair cat was referred for investigation of lethargy, weight loss, pyrexia and upper respiratory tract signs. On computed tomography, an expansile, osteodestructive lesion in the right tympanic bulla was identified. A soft tissue mass extended from the bulla into the nasopharynx, cranium and subcutaneous tissues. The nasopharyngeal mass ruptured during handling, liberating purulent material from which Pasteurella multocida was isolated in pure culture. The lesion was most likely an atypical, abscessated nasopharyngeal polyp. The cat was treated with bulla osteotomy and antibiotics, and made a complete recovery.
Collapse
Affiliation(s)
- Joanna L Pilton
- Faculty of Veterinary Science, University of Sydney, Sydney, Australia
| | - Charles J Ley
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Katja Voss
- Faculty of Veterinary Science, University of Sydney, Sydney, Australia
| | | | - Vanessa R Barrs
- Faculty of Veterinary Science, University of Sydney, Sydney, Australia
| | - Julia A Beatty
- Faculty of Veterinary Science, University of Sydney, Sydney, Australia
| |
Collapse
|
20
|
Kondo A, Tokuda H, Matsushima-Nishiwaki R, Kuroyanagi G, Yamamoto N, Mizutani J, Kozawa O, Otsuka T. Rho-kinase limits BMP-4-stimulated osteocalcin synthesis in osteoblasts: regulation of the p38 MAP kinase pathway. Life Sci 2013; 96:18-25. [PMID: 24368139 DOI: 10.1016/j.lfs.2013.12.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 12/03/2013] [Accepted: 12/11/2013] [Indexed: 12/13/2022]
Abstract
AIM We previously reported that bone morphogenetic protein-4 (BMP-4) stimulates the synthesis of osteocalcin via p38 mitogen-activated protein (MAP) kinase in osteoblast-like MC3T3-E1 cells, whereas p44/p42 MAP kinase plays as a negative regulator in the synthesis. In the present study, we investigated whether Rho-kinase is involved in BMP-4-stimulated osteocalcin synthesis in MC3T3-E1 cells. MAIN METHODS The levels of osteocalcin were measured by ELISA. The phosphorylation of each protein kinase was analyzed by Western blotting. The mRNA levels of osteocalcin were determined by real-time RT-PCR. KEY FINDINGS BMP-4 induced the phosphorylation of myosin phosphatase targeting subunit-1 (MYPT-1), a substrate of Rho-kinase. Y27632 or fasudil, specific inhibitors of Rho-kinase, which attenuated the MYPT-1 phosphorylation, significantly amplified the BMP-4-stimulated osteocalcin synthesis in a dose-dependent manner. The osteocalcin mRNA expression levels induced by BMP-4 were enhanced by Y27632 or fasudil. BMP-4-stimulated osteocalcin release was significantly up-regulated in Rho-knocked down cells with Rho A-siRNA. Y27632 or fasudil failed to affect the BMP-4-induced phosphorylation of SMAD1 or p44/p42 MAP kinase. On the other hand, Y27632 or fasudil markedly strengthened the phosphorylation levels of p38 MAP kinase induced by BMP-4. SIGNIFICANCE These results strongly suggest that Rho-kinase negatively regulates BMP-4-stimulated osteocalcin synthesis via the p38 MAP kinase pathway in osteoblasts.
Collapse
Affiliation(s)
- Akira Kondo
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Haruhiko Tokuda
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan; Department of Clinical Laboratory, National Center for Geriatrics and Gerontology, Obu, Aichi 474-8511, Japan.
| | | | - Gen Kuroyanagi
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Naohiro Yamamoto
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Jun Mizutani
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | - Osamu Kozawa
- Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194, Japan
| | - Takanobu Otsuka
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| |
Collapse
|
21
|
Kubatzky KF, Kloos B, Hildebrand D. Signaling cascades of Pasteurella multocida toxin in immune evasion. Toxins (Basel) 2013; 5:1664-81. [PMID: 24064721 PMCID: PMC3798879 DOI: 10.3390/toxins5091664] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 09/17/2013] [Accepted: 09/17/2013] [Indexed: 12/11/2022] Open
Abstract
Pasteurella multocida toxin (PMT) is a protein toxin found in toxigenic strains of Pasteurella multocida. PMT is the causative agent for atrophic rhinitis in pigs, a disease characterized by loss of nasal turbinate bones due to an inhibition of osteoblast function and an increase in osteoclast activity and numbers. Apart from this, PMT acts as a strong mitogen, protects from apoptosis and has an impact on the differentiation and function of immune cells. Many signaling pathways have been elucidated, however, the effect of these signaling cascades as a means to subvert the host’s immune system are just beginning to unravel.
Collapse
Affiliation(s)
- Katharina F Kubatzky
- Medical Microbiology and Hygiene, Department of Infectious Diseases, University Hospital Heidelberg, Im Neuenheimer Feld 324, Heidelberg 69120, Germany.
| | | | | |
Collapse
|
22
|
Abstract
In a world where most emerging and reemerging infectious diseases are zoonotic in nature and our contacts with both domestic and wild animals abound, there is growing awareness of the potential for human acquisition of animal diseases. Like other Pasteurellaceae, Pasteurella species are highly prevalent among animal populations, where they are often found as part of the normal microbiota of the oral, nasopharyngeal, and upper respiratory tracts. Many Pasteurella species are opportunistic pathogens that can cause endemic disease and are associated increasingly with epizootic outbreaks. Zoonotic transmission to humans usually occurs through animal bites or contact with nasal secretions, with P. multocida being the most prevalent isolate observed in human infections. Here we review recent comparative genomics and molecular pathogenesis studies that have advanced our understanding of the multiple virulence mechanisms employed by Pasteurella species to establish acute and chronic infections. We also summarize efforts being explored to enhance our ability to rapidly and accurately identify and distinguish among clinical isolates and to control pasteurellosis by improved development of new vaccines and treatment regimens.
Collapse
Affiliation(s)
- Brenda A Wilson
- Department of Microbiology and Host-Microbe Systems Theme of the Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.
| | | |
Collapse
|
23
|
Siegert P, Schmidt G, Papatheodorou P, Wieland T, Aktories K, Orth JHC. Pasteurella multocida toxin prevents osteoblast differentiation by transactivation of the MAP-kinase cascade via the Gα(q/11)--p63RhoGEF--RhoA axis. PLoS Pathog 2013; 9:e1003385. [PMID: 23696743 PMCID: PMC3656108 DOI: 10.1371/journal.ppat.1003385] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 04/11/2013] [Indexed: 11/19/2022] Open
Abstract
The 146-kDa Pasteurella multocida toxin (PMT) is the main virulence factor to induce P. multocida-associated progressive atrophic rhinitis in various animals. PMT leads to a destruction of nasal turbinate bones implicating an effect of the toxin on osteoblasts and/or osteoclasts. The toxin induces constitutive activation of Gα proteins of the Gq/11-, G12/13- and Gi-family by deamidating an essential glutamine residue. To study the PMT effect on bone cells, we used primary osteoblasts derived from rat calvariae and stromal ST-2 cells as differentiation model. As marker of functional osteoblasts the expression and activity of alkaline phosphatase, formation of mineralization nodules or expression of specific transcription factors as osterix was determined. Here, we show that the toxin inhibits differentiation and/or function of osteoblasts by activation of Gαq/11. Subsequently, Gαq/11 activates RhoA via p63RhoGEF, which specifically interacts with Gαq/11 but not with other G proteins like Gα12/13 and Gαi. Activated RhoA transactivates the mitogen-activated protein (MAP) kinase cascade via Rho kinase, involving Ras, MEK and ERK, resulting in inhibition of osteoblast differentiation. PMT-induced inhibition of differentiation was selective for the osteoblast lineage as adipocyte-like differentiation of ST-2 cells was not hampered. The present work provides novel insights, how the bacterial toxin PMT can control osteoblastic development by activating heterotrimeric G proteins of the Gαq/11-family and is a molecular pathogenetic basis for understanding the role of the toxin in bone loss during progressive atrophic rhinitis induced by Pasteurella multocida. Pasteurella multocida causes as a facultative pathogen various diseases in men and animals. One induced syndrome is atrophic rhinitis, which is a form of osteopenia, mainly characterized by facial distortion due to degradation of nasal turbinate bones. Strains, which especially affect bone tissue, produce the protein toxin P. multocida toxin (PMT). Importantly, PMT alone is capable to induce all symptoms of atrophic rhinitis. To cause osteopenia PMT influences the development and/or activity of specialized bone cells like osteoblasts and osteoclasts. Recently, we could identify the molecular mechanism of PMT. The toxin constitutively activates certain heterotrimeric G proteins by deamidation. Here, we studied the effect of PMT on the differentiation of osteoblasts. We demonstrate the direct action of PMT on osteoblasts and osteoblast-like cells and as a consequence inhibition of osteoblastic differentiation. Moreover, we revealed the underlying signal transduction pathway to impair proper osteoblast development. We show that PMT activates small GTPases in a Gαq/11 dependent manner via a non-ubiquitously expressed RhoGEF. In turn the mitogen-activated protein kinase pathway is transactivated leading to inhibition of osteoblastogenesis. Our findings present a mechanism how PMT hijacks host cell signaling pathways to hinder osteoblast development, which contributes to the syndrome of atrophic rhinitis.
Collapse
Affiliation(s)
- Peter Siegert
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- Hermann–Staudinger–Graduiertenschule Universität Freiburg, Freiburg, Germany
| | - Gudula Schmidt
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Panagiotis Papatheodorou
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Klaus Aktories
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, Universität Freiburg, Freiburg, Germany
- * E-mail: (KA); (JO)
| | - Joachim H. C. Orth
- Institut für Experimentelle und Klinische Pharmakologie und Toxikologie, Albert-Ludwigs-Universität Freiburg, Freiburg, Germany
- * E-mail: (KA); (JO)
| |
Collapse
|
24
|
Rho-kinase negatively regulates thyroid hormone-stimulated osteocalcin synthesis in osteoblasts. Biochimie 2013; 95:719-24. [DOI: 10.1016/j.biochi.2012.10.020] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Accepted: 10/25/2012] [Indexed: 11/18/2022]
|
25
|
Prowse PDH, Elliott CG, Hutter J, Hamilton DW. Inhibition of Rac and ROCK signalling influence osteoblast adhesion, differentiation and mineralization on titanium topographies. PLoS One 2013; 8:e58898. [PMID: 23505566 PMCID: PMC3591363 DOI: 10.1371/journal.pone.0058898] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Accepted: 02/08/2013] [Indexed: 11/18/2022] Open
Abstract
Reducing the time required for initial integration of bone-contacting implants with host tissues would be of great clinical significance. Changes in osteoblast adhesion formation and reorganization of the F-actin cytoskeleton in response to altered topography are known to be upstream of osteoblast differentiation, and these processes are regulated by the Rho GTPases. Rac and RhoA (through Rho Kinase (ROCK)). Using pharmacological inhibitors, we tested how inhibition of Rac and ROCK influenced osteoblast adhesion, differentiation and mineralization on PT (Pre-treated) and SLA (sandblasted large grit, acid etched) topographies. Inhibition of ROCK, but not Rac, significantly reduced adhesion number and size on PT, with adhesion size consistent with focal complexes. After 1 day, ROCK, but not Rac inhibition increased osteocalcin mRNA levels on SLA and PT, with levels further increasing at 7 days post seeding. ROCK inhibition also significantly increased bone sialoprotein expression at 7 days, but not BMP-2 levels. Rac inhibition significantly reduced BMP-2 mRNA levels. ROCK inhibition increased nuclear translocation of Runx2 independent of surface roughness. Mineralization of osteoblast cultures was greater on SLA than on PT, but was increased by ROCK inhibition and attenuated by Rac inhibition on both topographies. In conclusion, inhibition of ROCK signalling significantly increases osteoblast differentiation and biomineralization in a topographic dependent manner, and its pharmacological inhibition could represent a new therapeutic to speed bone formation around implanted metals and in regenerative medicine applications.
Collapse
Affiliation(s)
- Paul D. H. Prowse
- Department of Anatomy, Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Christopher G. Elliott
- Department of Cell Biology, Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
| | - Jeff Hutter
- Department of Physics and Astronomy, The University of Western Ontario, London, Ontario, Canada
| | - Douglas W. Hamilton
- Department of Anatomy, Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
- Department of Cell Biology, Graduate Program of Biomedical Engineering, The University of Western Ontario, London, Ontario, Canada
- Division of Oral Biology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada
- * E-mail:
| |
Collapse
|
26
|
Satija NK, Sharma D, Afrin F, Tripathi RP, Gangenahalli G. High throughput transcriptome profiling of lithium stimulated human mesenchymal stem cells reveals priming towards osteoblastic lineage. PLoS One 2013; 8:e55769. [PMID: 23383279 PMCID: PMC3559497 DOI: 10.1371/journal.pone.0055769] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2012] [Accepted: 01/04/2013] [Indexed: 02/07/2023] Open
Abstract
Human mesenchymal stem cells (hMSCs) present in the bone marrow are the precursors of osteoblasts, chondrocytes and adipocytes, and hold tremendous potential for osteoregenerative therapy. However, achieving directed differentiation into osteoblasts has been a major concern. The use of lithium for enhancing osteogenic differentiation has been documented in animal models but its effect in humans is not clear. We, therefore, performed high throughput transcriptome analysis of lithium-treated hMSCs to identify altered gene expression and its relevance to osteogenic differentiation. Our results show suppression of proliferation and enhancement of alkaline phosphatase (ALP) activity upon lithium treatment of hMSCs under non-osteogenic conditions. Microarray profiling of lithium-stimulated hMSC revealed decreased expression of adipogenic genes (CEBPA, CMKLR1, HSD11B1) and genes involved in lipid biosynthesis. Interestingly, osteoclastogenic factors and immune responsive genes (IL7, IL8, CXCL1, CXCL12, CCL20) were also downregulated. Negative transcriptional regulators of the osteogenic program (TWIST1 and PBX1) were suppressed while genes involved in mineralization like CLEC3B and ATF4 were induced. Gene ontology analysis revealed enrichment of upregulated genes related to mesenchymal cell differentiation and signal transduction. Lithium priming led to enhanced collagen 1 synthesis and osteogenic induction of lithium pretreated MSCs resulted in enhanced expression of Runx2, ALP and bone sialoprotein. However, siRNA-mediated knockdown of RRAD, CLEC3B and ATF4 attenuated lithium-induced osteogenic priming, identifying a role for RRAD, a member of small GTP binding protein family, in osteoblast differentiation. In conclusion, our data highlight the transcriptome reprogramming potential of lithium resulting in higher propensity of lithium "primed" MSCs for osteoblastic differentiation.
Collapse
Affiliation(s)
- Neeraj Kumar Satija
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| | - Deepa Sharma
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| | - Farhat Afrin
- Department of Biotechnology, Hamdard University, Hamdard Nagar, New Delhi, India
| | - Rajendra P. Tripathi
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| | - Gurudutta Gangenahalli
- Stem Cell & Gene Therapy Research Group, Institute of Nuclear Medicine & Allied Sciences, Brig. S K Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
27
|
Agas D, Marchetti L, Hurley MM, Sabbieti MG. Prostaglandin F2α: a bone remodeling mediator. J Cell Physiol 2012; 228:25-9. [PMID: 22585670 DOI: 10.1002/jcp.24117] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Prostaglandin F2α (PGF2α) plays multiple roles on bone metabolism by regulating a wide range of signaling pathways. PGF2α, via activation of PKC, stimulates Na-dependent inorganic phosphate (Pi) transport system in osteoblasts; up-regulates interleukin (IL)-6 synthesis; increases vascular endothelial growth factor (VEGF). In addition, PGF2α acts as a strong mitogenic and survival agent on osteoblasts, and these effects are, at least in part, mediated by the binding of fibroblast growth factor-2 (FGF-2) to the specific receptor FGFR1. The understanding of PGF2α intracellular network, albeit complex to clarify, provides molecular bases useful to identify the players of osteoblast proliferation, apoptosis, and the associated angiogenic processes. Indeed, the molecular mechanism that underline PGF2α-regulated bone metabolism may be a promising platform for the development of novel targeted therapies in the treatment of bone disorders and disease.
Collapse
Affiliation(s)
- Dimitrios Agas
- School of Biosciences and Biotechnology, University of Camerino, Camerino (MC), Italy
| | | | | | | |
Collapse
|
28
|
Bannai Y, Aminova LR, Faulkner MJ, Ho M, Wilson BA. Rho/ROCK-dependent inhibition of 3T3-L1 adipogenesis by G-protein-deamidating dermonecrotic toxins: differential regulation of Notch1, Pref1/Dlk1, and β-catenin signaling. Front Cell Infect Microbiol 2012; 2:80. [PMID: 22919671 PMCID: PMC3417509 DOI: 10.3389/fcimb.2012.00080] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Accepted: 05/22/2012] [Indexed: 01/11/2023] Open
Abstract
The dermonecrotic toxins from Pasteurella multocida (PMT), Bordetella (DNT), Escherichia coli (CNF1-3), and Yersinia (CNFY) modulate their G-protein targets through deamidation and/or transglutamination of an active site Gln residue, which results in activation of the G protein and its cognate downstream signaling pathways. Whereas DNT and the CNFs act on small Rho GTPases, PMT acts on the α subunit of heterotrimeric Gq, Gi, and G12/13 proteins. We previously demonstrated that PMT potently blocks adipogenesis and adipocyte differentiation in a calcineurin-independent manner through downregulation of Notch1 and stabilization of β-catenin and Pref1/Dlk1, key proteins in signaling pathways strongly linked to cell fate decisions, including fat and bone development. Here, we report that similar to PMT, DNT, and CNF1 completely block adipogenesis and adipocyte differentiation by preventing upregulation of adipocyte markers, PPARγ and C/EBPα, while stabilizing the expression of Pref1/Dlk1 and β-catenin. We show that the Rho/ROCK inhibitor Y-27632 prevented or reversed these toxin-mediated effects, strongly supporting a role for Rho/ROCK signaling in dermonecrotic toxin-mediated inhibition of adipogenesis and adipocyte differentiation. Toxin treatment was also accompanied by downregulation of Notch1 expression, although this inhibition was independent of Rho/ROCK signaling. We further show that PMT-mediated downregulation of Notch1 expression occurs primarily through G12/13 signaling. Our results reveal new details of the pathways involved in dermonecrotic toxin action on adipocyte differentiation, and the role of Rho/ROCK signaling in mediating toxin effects on Wnt/β-catenin and Notch1 signaling, and in particular the role of Gq and G12/13 in mediating PMT effects on Rho/ROCK and Notch1 signaling.
Collapse
Affiliation(s)
- Yuka Bannai
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana IL, USA
| | | | | | | | | |
Collapse
|
29
|
Taylor RM, Kashima TG, Hemingway FKE, Dongre A, Knowles HJ, Athanasou NA. CD14- mononuclear stromal cells support (CD14+) monocyte-osteoclast differentiation in aneurysmal bone cyst. J Transl Med 2012; 92:600-5. [PMID: 22330339 DOI: 10.1038/labinvest.2012.5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Aneurysmal bone cyst (ABC) is a benign osteolytic bone lesion in which there are blood-filled spaces separated by fibrous septa containing giant cells. The nature of the giant cells in this lesion and the mechanism of bone destruction in ABC is not certain. In this study, we have analysed several characteristics of mononuclear and multinucleated cells in the ABC and examined the cellular and molecular mechanisms of ABC osteolysis. The antigenic and functional phenotype of giant cells in ABC was determined by histochemistry/immunohistochemistry using antibodies to macrophage and osteoclast markers. Giant cells and CD14+ and CD14- mononuclear cells were isolated from ABC specimens and cultured on dentine slices and coverslips with receptor activator of nuclear factor κB ligand (RANKL)+/- macrophage-colony stimulating factor (M-CSF) and functional and cytochemical evidence of osteoclast differentiation sought. Giant cells in ABC expressed an osteoclast-like phenotype (CD51+, CD14-, cathepsin K+, TRAP+) and were capable of lacunar resorption, which was inhibited by zoledronate, calcitonin and osteoprotegerin (OPG). When cultured with RANKL±M-CSF, CD14+, but not CD14-, mononuclear cells differentiated into TRAP+ multinucleated cells that were capable of lacunar resorption. M-CSF was not necessary for osteoclast formation from CD14+ cell cultures. CD14- cells variably expressed RANKL, OPG and M-CSF but supported osteoclast differentiation. Our findings show that the giant cells in ABC express an osteoclast-like phenotype and are formed from CD14+ macrophage precursors. CD14- mononuclear stromal cells express osteoclastogenic factors and most likely interact with CD14+ cells to form osteoclast-like giant cells by a RANKL-dependent mechanism.
Collapse
Affiliation(s)
- Richard Mi Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Nuffield Orthopaedic Centre, Oxford, UK
| | | | | | | | | | | |
Collapse
|
30
|
Lax A. The Pasteurella multocida toxin: a new paradigm for the link between bacterial infection and cancer. Curr Top Microbiol Immunol 2012; 361:131-44. [PMID: 22695919 DOI: 10.1007/82_2012_236] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2022]
Abstract
The concept that bacterial infection could cause cancer has only recently become accepted because of the strong epidemiological and molecular evidence for a major carcinogenic role played by Helicobacter pylori. However, information on other potential bacterial carcinogens is very limited and thereby unconvincing. A different approach is to assess bacteria for potentially pro-carcinogenic properties. The Pasteurella multocida toxin (PMT) has many properties that mark it out as a potential carcinogen. PMT is a highly potent mitogen and has been demonstrated to block apoptosis. PMT modifies and activates members of three of the four families of heterotrimeric G-proteins, all of which have potential roles in carcinogenesis. Many signalling components downstream of these G-proteins are known proto-oncogenes and have been shown to be activated by PMT. These include, amongst others, the Rho GTPase, focal adhesion kinase, cyclooxygenase-2, β-catenin signalling and calcium signalling. PMT action potentially influences many of the acquired Hanahan/Weinberg capabilities necessary for oncogenic transformation. Although there is little evidence that PMT might have a role in human cancer, it serves as an important and novel paradigm for a bacterial link to cancer.
Collapse
Affiliation(s)
- Alistair Lax
- Department of Microbiology, King's College London Dental Institute, London, UK.
| |
Collapse
|
31
|
Swine atrophic rhinitis caused by pasteurella multocida toxin and bordetella dermonecrotic toxin. Curr Top Microbiol Immunol 2012; 361:113-29. [PMID: 22411430 DOI: 10.1007/82_2012_206] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
Atrophic rhinitis is a widespread and economically important swine disease caused by Pasteurella multocida and Bordetella bronchiseptica. The disease is characterized by atrophy of the nasal turbinate bones, which results in a shortened and deformed snout in severe cases. P. multocida toxin and B. bronchiseptica dermonecrotic toxin have been considered to independently or cooperatively disturb the osteogenesis of the turbinate bone by inhibiting osteoblastic differentiation and/or stimulating bone resorption by osteoclasts. Recently, the intracellular targets and molecular actions of both toxins have been clarified, enabling speculation on the intracellular signals leading to the inhibition of osteogenesis.
Collapse
|
32
|
Pasteurella multocida toxin interaction with host cells: entry and cellular effects. Curr Top Microbiol Immunol 2012; 361:93-111. [PMID: 22552700 PMCID: PMC4408768 DOI: 10.1007/82_2012_219] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
The mitogenic dermonecrotic toxin from Pasteurella multocida (PMT) is a 1285-residue multipartite protein that belongs to the A-B family of bacterial protein toxins. Through its G-protein-deamidating activity on the α subunits of heterotrimeric G(q)-, G(i)- and G(12/13)-proteins, PMT potently stimulates downstream mitogenic, calcium, and cytoskeletal signaling pathways. These activities lead to pleiotropic effects in different cell types, which ultimately result in cellular proliferation, while inhibiting cellular differentiation, and account for the myriad of physiological outcomes observed during infection with toxinogenic strains of P. multocida.
Collapse
|
33
|
Abstract
The mitogenic toxin from Pasteurella multocida (PMT) is a member of the dermonecrotic toxin family, which includes toxins from Bordetella, Escherichia coli and Yersinia. Members of the dermonecrotic toxin family modulate G-protein targets in host cells through selective deamidation and/or transglutamination of a critical active site Gln residue in the G-protein target, which results in the activation of intrinsic GTPase activity. Structural and biochemical data point to the uniqueness of PMT among these toxins in its structure and action. Whereas the other dermonecrotic toxins act on small Rho GTPases, PMT acts on the α subunits of heterotrimeric G(q) -, G(i) - and G(12/13) -protein families. To date, experimental evidence supports a model in which PMT potently stimulates various mitogenic and survival pathways through the activation of G(q) and G(12/13) signaling, ultimately leading to cellular proliferation, whilst strongly inhibiting pathways involved in cellular differentiation through the activation of G(i) signaling. The resulting cellular outcomes account for the global physiological effects observed during infection with toxinogenic P. multocida, and hint at potential long-term sequelae that may result from PMT exposure.
Collapse
Affiliation(s)
- Brenda A Wilson
- Department of Microbiology and Host-Microbe Systems Theme of the Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | | |
Collapse
|
34
|
Rho kinase inhibitors stimulate the migration of human cultured osteoblastic cells by regulating actomyosin activity. Cell Mol Biol Lett 2011; 16:279-95. [PMID: 21394446 PMCID: PMC6275969 DOI: 10.2478/s11658-011-0006-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 03/02/2011] [Indexed: 11/20/2022] Open
Abstract
We investigated the effects of Rho-associated kinase (ROCK) on migration and cytoskeletal organization in primary human osteoblasts and Saos-2 human osteosarcoma cells. Both cell types were exposed to two different ROCK inhibitors, Y-27632 and HA-1077. In the improved motility assay used in the present study, Y-27632 and HA-1077 significantly increased the migration of both osteoblasts and osteosarcoma cells on plastic in a dose-dependent and reversible manner. Fluorescent images showed that cells of both types cultured with Y-27632 or HA-1077 exhibited a stellate appearance, with poor assembly of stress fibers and focal contacts. Western blotting showed that ROCK inhibitors reduced myosin light chain (MLC) phosphorylation within 5 min without affecting overall myosin light-chain protein levels. Inhibition of ROCK activity is thought to enhance the migration of human osteoblasts through reorganization of the actin cytoskeleton and regulation of myosin activity. ROCK inhibitors may be potentially useful as anabolic agents to enhance the biocompatibility of bone and joint prostheses.
Collapse
|
35
|
Sims NA. EPHs and ephrins: Many pathways to regulate osteoblasts and osteoclasts. ACTA ACUST UNITED AC 2010. [DOI: 10.1138/20100463] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
36
|
Chen NX, Chen X, O'Neill KD, Atkinson SJ, Moe SM. RhoA/Rho kinase (ROCK) alters fetuin-A uptake and regulates calcification in bovine vascular smooth muscle cells (BVSMC). Am J Physiol Renal Physiol 2010; 299:F674-80. [PMID: 20610533 DOI: 10.1152/ajprenal.00730.2009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
RhoA/Rho kinases (ROCK) play a critical role in vascular smooth muscle cell (VSMC) actin cytoskeleton organization, differentiation, and function and are implicated in the pathogenesis of cardiovascular disease. We have previously determined that an important step in the regulation of calcification is fetuin-A endocytosis, a process that is dependent on changes in the cytoskeleton, which, in turn, is known to be affected by the RhoA/ROCK signaling pathway. In the present study, bovine VSMC (BVSMC) were treated with the ROCK inhibitor Y-27632 or transfected with ROCK small interfering (si) RNA to knock down ROCK expression. Both conditions resulted in reduced actin stress fibers and increased Cy5-labeled fetuin-A uptake. Inhibition of ROCK by Y-27632 or siRNA also significantly increased BVSMC alkaline phosphatase (ALP) activity and calcification of BVSMC and rat aorta organ cultures. Cells were then incubated in calcification media in the presence or absence of Y-27632 and matrix vesicles (MV) isolated by collagenase digestion. These MV, isolated from BVSMC incubated with Y-27632, had increased ALP activity and increased ability of MV to subsequently calcify collagen by 66%. In contrast, activation of RhoA, which is upstream of ROCK, by transfecting plasmids encoding the dominant active Rho GTPase mutant (Rho-L63) led to decreased fetuin-A uptake and reduced calcification in BVSMC. These results demonstrate that the RhoA/ROCK signaling pathway is an important negative regulator of vascular calcification.
Collapse
Affiliation(s)
- Neal X Chen
- Indiana Univ. School of Medicine, Indianapolis, IN 46202, USA.
| | | | | | | | | |
Collapse
|
37
|
Yotsumoto N, Takeoka M, Yokoyama M. Tail-suspended mice lacking calponin H1 experience decreased bone loss. TOHOKU J EXP MED 2010; 221:221-7. [PMID: 20551601 DOI: 10.1620/tjem.221.221] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Calponin h1 (CNh1) is an actin-binding protein originally isolated from vascular smooth muscle and has been reported to suppress bone formation. We are therefore curious how CNh1 is involved in bone loss that is caused by space flight in microgravity. We assessed the effects of tail suspension (TS) in C57BL/6J wild (CN+/+) and CNh1-deleted (CN-/-) mice to elucidate the role of CNh1 in bone loss under weightless conditions. Bone mineral density (BMD) of tibiae was measured by single energy X-ray absorptiometry, and bone volume fraction (BV/TV), mineral apposition rate (MAR), and bone formation rate (BFR/BS) were measured by bone histomorphometry. BMD, BV/TV, MAR, and BFR/BS were lower in CN+/+ mice with TS than in those without. In the CN-/- group, however, the decrease in each of these parameters by TS was ameliorated. Decreases in serum osteocalcin levels by TS in CN+/+ mice were attenuated in CN-/- mice. Furthermore, urinary deoxypyridinolin (DPD), an indicator of bone resorption, was increased in CN+/+ mice following TS, but not in CN-/- mice. In transfection experiments, the degree of induction of bone formation markers, alkaline phosphatase (ALP) activity and bone morphogenetic protein (BMP)-4 mRNA expression, under stimulation with BMP-2, was lower in MC3T3-E1 mouse osteoblast-like cells expressing CNh1 than that in mock transfected cells. Notably, the BMP-2-induced ALP activity was decreased by CNh1 expression, which was partially rescued by treatment with the Rho kinase inhibitor Y27632. Taken together, these results indicate that CNh1 is responsible for weightlessness-induced bone loss in part through Rho signaling pathway.
Collapse
|
38
|
Kato K, Tokuda H, Natsume H, Adachi S, Matsushima-Nishiwaki R, Minamitani C, Mizutani J, Kozawa O, Otsuka T. Rho-kinase regulates prostaglandin D(2)-stimulated heat shock protein 27 induction in osteoblasts. Exp Ther Med 2010; 1:579-583. [PMID: 22993579 DOI: 10.3892/etm_00000091] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2010] [Accepted: 05/18/2010] [Indexed: 11/05/2022] Open
Abstract
We previously reported that prostaglandin D(2) (PGD(2)) stimulates heat shock protein 27 (HSP27) induction through p44/p42 mitogen-activated protein (MAP) kinase, p38 MAP kinase and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in osteoblast-like MC3T3-E1 cells. In addition, we recently showed that PGD(2) activates Rho-kinase, resulting in the regulation of interleukin-6 synthesis via activation of p38 MAP kinase but not p44/p42 MAP kinase in these cells. In the present study, in order to investigate whether Rho-kinase is involved in the PGD(2)-stimulated HSP27 induction in MC3T3-E1 cells, we examined the effects of Rho-kinase inhibitors on HSP27 induction. Y27632 and fasudil, Rho-kinase inhibitors, markedly suppressed the HSP27 induction stimulated by PGD(2) in a dose-dependent manner without affecting levels of HSP70 in the presence of PGD(2). Immunofluorescence microscopy studies also revealed that Y27632 and fasudil markedly suppressed the induction of HSP27. Y27632 and fasudil attenuated the PGD(2)-induced phosphorylation levels of SAPK/JNK. In conclusion, Rho-kinase inhibitors regulate PGD(2)-stimulated HSP27 induction via activation of both SAPK/JNK and p38 MAP kinase in osteoblasts.
Collapse
Affiliation(s)
- Kenji Kato
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601; ; Department of Pharmacology, Gifu University Graduate School of Medicine, Gifu 501-1194
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Neighbor of Brca1 gene (Nbr1) functions as a negative regulator of postnatal osteoblastic bone formation and p38 MAPK activity. Proc Natl Acad Sci U S A 2010; 107:12913-8. [PMID: 20616007 DOI: 10.1073/pnas.0913058107] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
The neighbor of Brca1 gene (Nbr1) functions as an autophagy receptor involved in targeting ubiquitinated proteins for degradation. It also has a dual role as a scaffold protein to regulate growth-factor receptor and downstream signaling pathways. We show that genetic truncation of murine Nbr1 leads to an age-dependent increase in bone mass and bone mineral density through increased osteoblast differentiation and activity. At 6 mo of age, despite normal body size, homozygous mutant animals (Nbr1(tr/tr)) have approximately 50% more bone than littermate controls. Truncated Nbr1 (trNbr1) co-localizes with p62, a structurally similar interacting scaffold protein, and the autophagosome marker LC3 in osteoblasts, but unlike the full-length protein, trNbr1 fails to complex with activated p38 MAPK. Nbr1(tr/tr) osteoblasts and osteoclasts show increased activation of p38 MAPK, and significantly, pharmacological inhibition of the p38 MAPK pathway in vitro abrogates the increased osteoblast differentiation of Nbr1(tr/tr) cells. Nbr1 truncation also leads to increased p62 protein expression. We show a role for Nbr1 in bone remodeling, where loss of function leads to perturbation of p62 levels and hyperactivation of p38 MAPK that favors osteoblastogenesis.
Collapse
|
40
|
Natsume H, Tokuda H, Adachi S, Takai S, Matsushima-Nishiwaki R, Kato K, Minamitani C, Niida S, Mizutani J, Kozawa O, Otsuka T. Rho-kinase limits FGF-2-stimulated VEGF release in osteoblasts. Bone 2010; 46:1068-74. [PMID: 20114091 DOI: 10.1016/j.bone.2010.01.378] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2009] [Revised: 12/16/2009] [Accepted: 01/11/2010] [Indexed: 11/19/2022]
Abstract
We previously reported that basic fibroblast growth factor (FGF-2) stimulates the release of vascular endothelial growth factor (VEGF) via p44/p42 mitogen-activated protein (MAP) kinase and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in osteoblast-like MC3T3-E1 cells and that FGF-2-activated p38 MAP kinase negatively regulates the VEGF release in osteoblast-like MC3T3-E1 cells. In the present study, we investigated whether Rho-kinase is involved in FGF-2-stimulated VEGF release in MC3T3-E1 cells. FGF-2 induced the phosphorylation of myosin phosphatase targeting subunit (MYPT-1), a substrate of Rho-kinase. Y27632, a specific inhibitor of Rho-kinase, which attenuated the MYPT-1 phosphorylation, significantly enhanced the FGF-2-stimulated VEGF release. Fasudil, another Rho-kinase inhibitor, also amplified the VEGF release. FGF-2 significantly stimulated VEGF accumulation and fasudil enhanced FGF-2-stimulated VEGF accumulation also in whole cell lysates. Neither Y27632 nor fasudil affected the phosphorylation levels of p44/p42 MAP kinase or p38 MAP kinase. Y27632 and fasudil markedly strengthened the FGF-2-induced phosphorylation of SAPK/JNK. Y27632 as well as fasudil enhanced FGF-2-stimulated VEGF release and Y27632 enhanced the FGF-2-induced phosphorylation levels of SAPK/JNK also in human osteoblasts. These results strongly suggest that Rho-kinase negatively regulates FGF-2-stimulated VEGF release in osteoblasts.
Collapse
Affiliation(s)
- Hideo Natsume
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
ZHAO J, MAO YJ, GU ZY. Progress in Researches of Eph/ephrin Signaling in Coupling of Bone Resorption and Bone Formation*. PROG BIOCHEM BIOPHYS 2010. [DOI: 10.3724/sp.j.1206.2009.00093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Kanazawa I, Yamaguchi T, Yano S, Yamauchi M, Sugimoto T. Fasudil hydrochloride induces osteoblastic differentiation of stromal cell lines, C3H10T1/2 and ST2, via bone morphogenetic protein-2 expression. Endocr J 2010; 57:415-21. [PMID: 20154408 DOI: 10.1507/endocrj.k09e-328] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Rho-kinase (ROK), downstream of the mevalonate pathway, is detrimental to vessels, and suppressing its activity is a target for the treatment of human disease such as coronary artery disease and pulmonary hypertension. Recent studies have shown that ROK has a crucial role in bone metabolism. However, the role of ROK in stromal cells is still unclear. The present study was undertaken to investigate the effect of a ROK inhibitor, fasudil hydrochloride, on stromal cell lines, C3H10T1/2 and ST2. In both cells, Fasudil significantly stimulated alkaline phosphatase activity and enhanced cell mineralization. Moreover, fasudil significantly increased the mRNA expression of collagen-I, osteocalcin, and bone morphogenetic protein-2 (BMP-2). Supplementation of noggin, a BMP-2 antagonist, significantly reversed the fasudil-induced collagen-I and osteocalcin mRNA expression in both cells. These findings suggest that fasudil induces the osteoblastic differentiation of stromal cells via enhancing BMP-2 expression, and that this drug might be beneficial for not only atherosclerosis but also osteoporosis by promoting bone formation.
Collapse
Affiliation(s)
- Ippei Kanazawa
- Department of Internal Medicine 1, Shimane University Faculty of Medicine, Izumo, Japan.
| | | | | | | | | |
Collapse
|
43
|
Involvement of Rho-kinase in prostaglandin E1-stimulated VEGF synthesis through stress-activated protein kinase/c-Jun N-terminal kinase in osteoblast-like MC3T3-E1 cells. Prostaglandins Other Lipid Mediat 2009; 90:1-6. [DOI: 10.1016/j.prostaglandins.2009.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2009] [Revised: 05/18/2009] [Accepted: 06/03/2009] [Indexed: 01/05/2023]
|
44
|
Kanazawa I, Yamaguchi T, Yano S, Yamauchi M, Sugimoto T. Activation of AMP kinase and inhibition of Rho kinase induce the mineralization of osteoblastic MC3T3-E1 cells through endothelial NOS and BMP-2 expression. Am J Physiol Endocrinol Metab 2009; 296:E139-46. [PMID: 19001547 DOI: 10.1152/ajpendo.90677.2008] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
AMP-activated protein kinase (AMPK) and Rho kinase (ROK) are known to modulate the mevalonate pathway. Activation of AMPK suppresses 3-hydroxy-3-methylglutaryl (HMG)-coenzyme A (CoA) reductase. ROK acts downstream of HMG-CoA reductase, and its inhibition exerts antiatherosclerosis effects. However, whether or not these enzymes are involved in bone metabolism is unclear. The present study was undertaken to investigate the effects of an AMPK activator, 5-aminoimidazole-4-carboxamide1-beta-d-ribonucleoside (AICAR), and a ROK inhibitor, fasudil hydrochrolide, on the mineralization of osteoblastic MC3T3-E1 cells. Real-time PCR and mineralization stainings revealed that both AICAR and fasudil significantly stimulated endothelial nitric oxide synthase (eNOS), bone morphogenetic protein-2 (BMP-2), and osteocalcin mRNA expression as well as mineralization in the cells. Supplementation of either mevalonate or geranyl-geranyl pyrophosphate, the downstream molecules of HMG-CoA reductase, or coincubation with either a nitric oxide synthase inhibitor, N(G)-nitro-l-arginine methyl ester, or a BMP-2 antagonist, noggin, significantly reversed these AICAR-induced reactions. Western blot analysis showed that AICAR activated protein kinase B and extracellular signal-regulated kinase (ERK). ERK inhibitor significantly reversed the AICAR-induced increase in eNOS and BMP-2 mRNA expression. Measurement of ROK activities by enzyme-linked immunosorbent assay revealed that both AICAR and fasudil significantly suppressed the phosphorylation of the myosin-binding subunit of myosin phosphate, a ROK substrate. These findings suggest that the AMPK activator and the ROK inhibitor are able to stimulate the mineralization of osteoblasts through modulating the mevalonate pathway. These agents could be candidate drugs that promote bone formation for the treatment of osteoporosis.
Collapse
Affiliation(s)
- Ippei Kanazawa
- Dept. of Internal Medicine 1, Shimane Univ. Faculty of Medicine, 89-1 Enya-cho, Izumo 693-8501, Japan
| | | | | | | | | |
Collapse
|
45
|
Matsuo K. Eph and ephrin interactions in bone. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 658:95-103. [PMID: 19950019 DOI: 10.1007/978-1-4419-1050-9_10] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bone cells communicate with each other using various cell surface molecules. Membrane-bound ephrin ligands and Eph tyrosine kinase receptors have been characterized in diverse biological processes, including angiogenesis and neuronal development. Several ephrins and Ephs are expressed in osteoclasts and osteoblasts and regulate bone mineral metabolism through bidirectional signaling into not only receptor-expressing cells but also into ligand-expressing cells. We propose that interaction between ephrinB2-expressing osteoclasts and EphB4-expressing osteoblasts facilitates the transition from bone resorption to bone formation during bone remodeling. Other groups have reported the regulation of ephrinB2 by PTH or PTHrP and the possible involvement of EphB4 in osteoarthritis. It is likely that various ephrins and Ephs mediate interaction among bone cells.
Collapse
Affiliation(s)
- Koichi Matsuo
- Collaborative Research Resources, School of Medicine, Keio University, 160-8582 Tokyo, Japan.
| |
Collapse
|
46
|
Rho-kinase inhibitors decrease TGF-beta-stimulated VEGF synthesis through stress-activated protein kinase/c-Jun N-terminal kinase in osteoblasts. Biochem Pharmacol 2008; 77:196-203. [PMID: 18996357 DOI: 10.1016/j.bcp.2008.10.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2008] [Revised: 10/09/2008] [Accepted: 10/09/2008] [Indexed: 11/20/2022]
Abstract
We have previously reported that transforming growth factor-beta (TGF-beta) stimulates the synthesis of vascular endothelial growth factor (VEGF) through p44/p42 mitogen-activated protein (MAP) kinase, p38 MAP kinase and stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) in osteoblast-like MC3T3-E1 cells. In order to investigate whether Rho-kinase is involved in the TGF-beta-stimulated VEGF synthesis in these cells we examined the effects of Rho-kinase inhibitors on the VEGF synthesis. TGF-beta time-dependently induced the phosphorylation of myosin phosphatase targeting subunit (MYPT-1) which is a well known substrate of Rho-kinase. Y27632 and fasudil, Rho-kinase inhibitors, significantly reduced the TGF-beta-stimulated VEGF synthesis as well as the MYPT-1 phosphorylation. Y27632 and fasudil failed to affect the TGF-beta-induced phosphorylation of p44/p42 MAP kinase, p38 MAP kinase or Smad2. On the contrary, Y27632 as well as fasudil markedly suppressed the TGF-beta-induced phosphorylation of SAPK/JNK. Taken together, our results strongly suggest that Rho-kinase regulates TGF-beta-stimulated VEGF synthesis via SAPK/JNK activation in osteoblasts.
Collapse
|
47
|
Minamitani C, Otsuka T, Takai S, Matsushima-Nishiwaki R, Adachi S, Hanai Y, Mizutani J, Tokuda H, Kozawa O. Involvement of Rho-kinase in prostaglandin F2alpha-stimulated interleukin-6 synthesis via p38 mitogen-activated protein kinase in osteoblasts. Mol Cell Endocrinol 2008; 291:27-32. [PMID: 18586382 DOI: 10.1016/j.mce.2008.05.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2007] [Revised: 04/24/2008] [Accepted: 05/19/2008] [Indexed: 11/18/2022]
Abstract
We have previously reported that prostaglandin F(2alpha) (PGF(2alpha)) stimulates interleukin-6 (IL-6), a potent bone resorptive agent, through p44/p42 mitogen-activated protein (MAP) kinase in osteoblast-like MC3T3-E1 cells. In the present study, we investigated whether Rho-kinase is implicated in the PGF(2alpha)-stimulated IL-6 synthesis in MC3T3-E1 cells. PGF(2alpha) time-dependently induced the phosphorylation of myosin phosphatase targeting subunit (MYPT-1), a Rho-kinase substrate. Y27632, a specific Rho-kinase inhibitor, significantly reduced the PGF(2alpha)-stimulated IL-6 synthesis as well as the MYPT-1 phosphorylation. Fasudil, another inhibitor of Rho-kinase, suppressed the PGF(2alpha)-stimulated IL-6 synthesis. Y27632 and fasudil failed to affect the PGF(2alpha)-induced phosphorylation of p44/p42 MAP kinase. SB203580 and BIRB0796, potent inhibitors of p38 MAP kinase, suppressed the IL-6 synthesis induced by PGF(2alpha). While SP600125, an inhibitor of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK), failed to reduce the synthesis. Y27632 as well as fasudil attenuated the PGF(2alpha)-induced phosphorylation of p38 MAP kinase. These results strongly suggest that Rho-kinase regulates PGF(2alpha)-stimulated IL-6 synthesis via p38 MAP kinase activation in osteoblasts.
Collapse
Affiliation(s)
- Chiho Minamitani
- Department of Orthopedic Surgery, Nagoya City University Graduate School of Medical Sciences, Nagoya 467-8601, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tokuda H, Takai S, Matsushima-Nishiwaki R, Hanai Y, Adachi S, Minamitani C, Mizutani J, Otsuka T, Kozawa O. Function of Rho-kinase in prostaglandin D2-induced interleukin-6 synthesis in osteoblasts. Prostaglandins Leukot Essent Fatty Acids 2008; 79:41-6. [PMID: 18771907 DOI: 10.1016/j.plefa.2008.07.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2008] [Revised: 06/27/2008] [Accepted: 07/10/2008] [Indexed: 11/24/2022]
Abstract
We have previously reported that prostaglandin D2 (PGD2) stimulates interleukin-6 (IL-6), a potent bone resorptive agent, in osteoblast-like MC3T3-E1 cells. In the present study, we investigated whether Rho-kinase is implicated in the PGD2-stimulated IL-6 synthesis in MC3T3-E1 cells. PGD2 time-dependently induced the phosphorylation of myosin phosphatase targeting subunit (MYPT-1), a Rho-kinase substrate. Y27632, a specific Rho-kinase inhibitor, significantly reduced the PGD2-stimulated IL-6 synthesis as well as the MYPT-1 phosphorylation. Fasudil, another inhibitor of Rho-kinase, suppressed the PGD2-stimulated IL-6 synthesis. The PGD2-stimulated IL-6 synthesis was reduced by PD98059, a MEK inhibitor, and SB203580, an inhibitor of p38 mitogen-activated protein (MAP) kinase, but not SP600125, an inhibitor of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK). However, Y27632 and fasudil failed to affect the PGD2-induced phosphorylation of p44/p42 MAP kinase. On the other hand, Y27632 as well as fasudil markedly attenuated the PGD2-induced phosphorylation of p38 MAP kinase. In addition, PGD2 additively induced IL-6 synthesis in combination with endothelin-1 which induces IL-6 synthesis through p38 MAP kinase regulated by Rho-kinase. These results strongly suggest that Rho-kinase regulates PGD2-stimulated IL-6 synthesis via p38 MAP kinase activation in osteoblasts.
Collapse
Affiliation(s)
- Haruhiko Tokuda
- Department of Clinical Laboratory, National Hospital for Geriatric Medicine, National Center for Geriatrics and Gerontology, 36-3 Gengo, Obu, Aichi 474-8511, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Intravesical botulinum toxin A injections plus hydrodistension can reduce nerve growth factor production and control bladder pain in interstitial cystitis: a molecular mechanism. Urology 2008; 72:463-4; author reply 464. [PMID: 18407329 DOI: 10.1016/j.urology.2008.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2008] [Revised: 02/08/2008] [Accepted: 02/08/2008] [Indexed: 11/23/2022]
|
50
|
Namazi H. Botulinum toxin type-A therapy in cluster headache: a novel molecular mechanism. J Headache Pain 2008; 9:133. [PMID: 18299799 PMCID: PMC3476182 DOI: 10.1007/s10194-008-0025-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2008] [Accepted: 02/04/2008] [Indexed: 11/28/2022] Open
Affiliation(s)
- Hamid Namazi
- Chamran Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|