1
|
Pieles O, Morsczeck C. The Role of Protein Kinase C During the Differentiation of Stem and Precursor Cells into Tissue Cells. Biomedicines 2024; 12:2735. [PMID: 39767642 PMCID: PMC11726769 DOI: 10.3390/biomedicines12122735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 01/05/2025] Open
Abstract
Protein kinase C (PKC) plays an essential role during many biological processes including development from early embryonic stages until the terminal differentiation of specialized cells. This review summarizes the current knowledge about the involvement of PKC in molecular processes during the differentiation of stem/precursor cells into tissue cells with a particular focus on osteogenic, adipogenic, chondrogenic and neuronal differentiation by using a comprehensive approach. Interestingly, studies examining the overall role of PKC, or one of its three isoform groups (classical, novel and atypical PKCs), often showed controversial results. A discrete observation of distinct isoforms demonstrated that the impact on differentiation differs highly between the isoforms, and that during a certain process, the influence of only some isoforms is crucial, while others are less important. In particular, PKCβ inhibits, and PKCδ strongly supports osteogenesis, whereas it is the other way around for adipogenesis. PKCε is another isoform that overwhelmingly supports adipogenic differentiation. In addition, PKCα plays an important role in chondrogenesis, while neuronal differentiation has been positively associated with numerous isoforms including classical, novel and atypical PKCs. In a cellular context, various upstream mediators, like the canonical and non-canonical Wnt pathways, endogenously control PKC activity and thus, their activity interferes with the influence of PKC on differentiation. Downstream of PKC, several proteins and pathways build the molecular bridge between the enzyme and the control of differentiation, of which only a few have been well characterized so far. In this context, PKC also cooperates with other kinases like Akt or protein kinase A (PKA). Furthermore, PKC is capable of directly phosphorylating transcription factors with pivotal function for a certain developmental process. Ultimately, profound knowledge about the role of distinct PKC isoforms and the involved signaling pathways during differentiation constitutes a promising tool to improve the use of stem cells in regenerative therapies by precisely manipulating the activity of PKC or downstream effectors.
Collapse
Affiliation(s)
| | - Christian Morsczeck
- Department of Oral and Maxillofacial Surgery, University Hospital Regensburg, Franz-Josef-Strauss-Allee 11, 93053 Regensburg, Germany;
| |
Collapse
|
2
|
Miao J, Tu Y, Jiang J, Ren R, Wu Q, Liang H, Wang T, Lin B, Wu J, Pan Y, Wang X, Jin H. VSIG4 inhibits RANKL-induced osteoclastogenesis by enhancing Nrf2-dependent antioxidant response against reactive oxygen species production. Int J Biol Macromol 2024; 260:129357. [PMID: 38216011 DOI: 10.1016/j.ijbiomac.2024.129357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 11/10/2023] [Accepted: 01/07/2024] [Indexed: 01/14/2024]
Abstract
Osteoporosis is a prevalent systemic skeletal disorder, particularly affecting postmenopausal women, primarily due to excessive production and activation of osteoclasts. However, the current anti-osteoporotic drugs utilized in clinical practice may lead to certain side effects. Therefore, it is necessary to further unravel the potential mechanisms regulating the osteoclast differentiation and to identify novel targets for osteoporosis treatment. This study revealed the most significant decline in VSIG4 expression among the VSIG family members. VSIG4 overexpression significantly inhibited RANKL-induced osteoclastogenesis and bone resorption function. Mechanistically, both western blot and immunofluorescence assay results demonstrated that VSIG4 overexpression attenuated the expression of osteoclast marker genes and dampened the activation of MAPK and NF-κB signaling pathways. Furthermore, VSIG4 overexpression could inhibit the generation of reactive oxygen species (ROS) and stimulate the expression of Nrf2 along with its downstream antioxidant enzymes via interaction with Keap1. Notably, a potent Nrf2 inhibitor, ML385, could reverse the inhibitory effect of VSIG4 on osteoclast differentiation. In line with these findings, VSIG4 overexpression also mitigated bone loss induced by OVX and attenuated the activation of osteoclasts in vivo. In conclusion, our results suggest that VSIG4 holds promise as a novel target for addressing postmenopausal osteoporosis. This is achieved by suppressing osteoclast formation via enhancing Nrf2-dependent antioxidant response against reactive oxygen species production.
Collapse
Affiliation(s)
- Jiansen Miao
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Yiting Tu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Junchen Jiang
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Rufeng Ren
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Qihang Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Haibo Liang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Tengjie Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Binghao Lin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Jingtao Wu
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China
| | - Youjin Pan
- The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China; Department of Endocrinology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China.
| | - Xiangyang Wang
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China.
| | - Haiming Jin
- Department of Orthopaedics, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000, China; The Second School of Medicine, Wenzhou Medical University, Wenzhou 325000, China.
| |
Collapse
|
3
|
TIAN L, SUN S, WANG J, LI W, WANG X. GINS2 affects activity/differentiation, apoptosis and proliferation of osteoblast and osteoclast in steroid-induced osteonecrosis of the femoral head by regulating P53/GADD45A signaling pathway. FOOD SCIENCE AND TECHNOLOGY 2022. [DOI: 10.1590/fst.09921] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Lei TIAN
- Shandong First Medical University, China
| | - Shui SUN
- Shandong First Medical University, China
| | - Jian WANG
- Shandong First Medical University, China
| | - Wei LI
- Shandong First Medical University, China
| | | |
Collapse
|
4
|
Mahajan S, Mellins ED, Faccio R. Diacylglycerol Kinase ζ Regulates Macrophage Responses in Juvenile Arthritis and Cytokine Storm Syndrome Mouse Models. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:137-146. [PMID: 31801815 PMCID: PMC6920556 DOI: 10.4049/jimmunol.1900721] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 11/04/2019] [Indexed: 12/18/2022]
Abstract
Dysregulation of monocyte and macrophage responses are often observed in children with systemic juvenile idiopathic arthritis (sJIA) and cytokine storm syndrome (CSS), a potentially fatal complication of chronic rheumatic diseases. Both conditions are associated with activation of TLR signaling in monocyte and macrophage lineage cells, leading to overwhelming inflammatory responses. Despite the importance of TLR engagement in activating proinflammatory macrophages, relatively little is known about activation of intrinsic negative regulatory pathways to attenuate excessive inflammatory responses. In this study, we demonstrate that loss of diacylglycerol (DAG) kinase (Dgk) ζ, an enzyme which converts DAG into phosphatidic acid, limits inflammatory cytokine production in an arthritic mouse model dependent on TLR2 signaling and in a CSS mouse model dependent on TLR9 signaling. In vitro, Dgkζ deficiency results in reduced production of TNF-α, IL-6, and IL-1β and in limited M1 macrophage polarization. Mechanistically, Dgkζ deficiency decreases STAT1 and STAT3 phosphorylation. Moreover, Dgkζ levels are increased in macrophages derived from mice with CSS or exposed to plasma from sJIA patients with active disease. Our data suggest that Dgkζ induction in arthritic conditions perpetuates systemic inflammatory responses mediated by macrophages and highlight a potential role of Dgkζ-DAG/phosphatidic acid axis as a modulator of inflammatory cytokine production in sJIA and CSS.
Collapse
Affiliation(s)
- Sahil Mahajan
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO 63110
| | - Elizabeth D Mellins
- Program in Immunology and Allergy, Department of Pediatrics, Stanford University, Stanford, CA 94305; and
| | - Roberta Faccio
- Department of Orthopedic Surgery, Musculoskeletal Research Center, Washington University School of Medicine, St. Louis, MO 63110;
- Shriners Hospitals for Children, St. Louis, MO 63110
| |
Collapse
|
5
|
Anesi A, Generali L, Sandoni L, Pozzi S, Grande A. From Osteoclast Differentiation to Osteonecrosis of the Jaw: Molecular and Clinical Insights. Int J Mol Sci 2019; 20:ijms20194925. [PMID: 31590328 PMCID: PMC6801843 DOI: 10.3390/ijms20194925] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 01/05/2023] Open
Abstract
Bone physiology relies on the delicate balance between resorption and formation of its tissue. Bone resorption depends on a process called osteoclastogenesis in which bone-resorbing cells, i.e., osteoclasts, are produced by the differentiation of more undifferentiated progenitors and precursors. This process is governed by two main factors, monocyte-colony stimulating factor (M-CSF) and receptor activator of NFκB ligand (RANKL). While the former exerts a proliferating effect on progenitors/precursors, the latter triggers a differentiation effect on more mature cells of the same lineage. Bone homeostasis requires a perfect space–time coordination of the involved signals. When osteoclastogenesis is poorly balanced with the differentiation of the bone forming counterparts, i.e., osteoblasts, physiological bone remodelling can turn into a pathological state, causing the systematic disruption of bone tissue which results in osteopenia or osteolysis. Examples of these conditions are represented by osteoporosis, Paget’s disease, bone metastasis, and multiple myeloma. Therefore, drugs targeting osteoclastogenesis, such as bisphosphonates and an anti-RANKL monoclonal antibody, have been developed and are currently used in the treatment of such diseases. Despite their demonstrated therapeutic efficacy, these agents are unfortunately not devoid of side effects. In this regard, a condition called osteonecrosis of the jaw (ONJ) has been recently correlated with anti-resorptive therapy. In this review we will address the involvement of osteoclasts and osteoclast-related factors in the pathogenesis of ONJ. It is to be hoped that a better understanding of the biological mechanisms underlying bone remodelling will help in the design a medical therapeutic approach for ONJ as an alternative to surgical procedures.
Collapse
Affiliation(s)
- Alexandre Anesi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy.
| | - Luigi Generali
- Department of Surgery, Medicine, Dentistry and Morphological Sciences with Transplant Surgery, Oncology and Regenerative Medicine Relevance, University of Modena and Reggio Emilia, 41121 Modena, Italy.
| | - Laura Sandoni
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125 Modena, Italy.
| | - Samantha Pozzi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Via del Pozzo 71, 41124 Modena, Italy.
| | - Alexis Grande
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via Giuseppe Campi 287, 41125 Modena, Italy.
| |
Collapse
|
6
|
Feng W, Li J, Liao S, Ma S, Li F, Zhong C, Li G, Wei Y, Huang H, Wei Q, Yao J, Liu Y. Gö6983 attenuates titanium particle-induced osteolysis and RANKL mediated osteoclastogenesis through the suppression of NFκB/JNK/p38 pathways. Biochem Biophys Res Commun 2018; 503:62-70. [PMID: 29856998 DOI: 10.1016/j.bbrc.2018.05.177] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Accepted: 05/26/2018] [Indexed: 10/14/2022]
Abstract
Osteoclast activation by wear particles has caused major difficulties for surgeons. Wear particles are the main causes of aseptic prosthetic loosening. Gö6983, a protein kinase C inhibitor, inhibits five subtypes of protein kinase C family members. Here, we found that Gö6983 had an obviously inhibitory effect on wear-particles-induced osteolysis in vivo. In vitro, Gö6983 inhibited RANKL-stimulated osteoclast formation and function by inhibiting the RANKL-stimulated nuclear factor-κB/JNK/p38 signaling pathway. We also observed that Go6983 had no effect on the differentiation of osteoblasts and osteoblast-associated genes expression. According to our data, Gö6983 has potential therapeutic effects for aseptic prosthetic loosening caused by osteoclast activation.
Collapse
Affiliation(s)
- Wenyu Feng
- Departments of Orthopedics, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Jia Li
- Departments of Pathology, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Shijie Liao
- Departments of Orthopedics, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Shiting Ma
- Departments of Orthopedics, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Feicui Li
- Departments of General Medicine, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Chaoyi Zhong
- Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Guodong Li
- Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China
| | - Yan Wei
- Departments of Pathology, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Huading Huang
- Departments of Cardiothoracic Surgery, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingjun Wei
- Departments of Orthopedics, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| | - Jun Yao
- Departments of Orthopedics, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| | - Yun Liu
- Departments of Orthopedics, The First Affliated Hospital of Guangxi Medical University, Nanning, Guangxi, China; Research Centre for Regenerative Medicine, Guangxi Key Laboratory of Regenerative Medicine, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
7
|
Zamani A, Decker C, Cremasco V, Hughes L, Novack DV, Faccio R. Diacylglycerol Kinase ζ (DGKζ) Is a Critical Regulator of Bone Homeostasis Via Modulation of c-Fos Levels in Osteoclasts. J Bone Miner Res 2015; 30:1852-63. [PMID: 25891971 PMCID: PMC4580562 DOI: 10.1002/jbmr.2533] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 04/07/2015] [Accepted: 04/10/2015] [Indexed: 12/31/2022]
Abstract
Increased diacylglycerol (DAG) levels are observed in numerous pathologies, including conditions associated with bone loss. However, the effects of DAG accumulation on the skeleton have never been directly examined. Because DAG is strictly controlled by tissue-specific diacylglycerol kinases (DGKs), we sought to examine the biological consequences of DAG accumulation on bone homeostasis by genetic deletion of DGKζ, a highly expressed DGK isoform in osteoclasts (OCs). Strikingly, DGKζ(-/-) mice are osteoporotic because of a marked increase in OC numbers. In vitro, DGKζ(-/-) bone marrow macrophages (BMMs) form more numerous, larger, and highly resorptive OCs. Surprisingly, although increased DAG levels do not alter receptor activator of NF-κB (RANK)/RANK ligand (RANKL) osteoclastogenic pathway, DGKζ deficiency increases responsiveness to the proliferative and pro-survival cytokine macrophage colony-stimulating factor (M-CSF). We find that M-CSF is responsible for increased DGKζ(-/-) OC differentiation by promoting higher expression of the transcription factor c-Fos, and c-Fos knockdown in DGKζ(-/-) cultures dose-dependently reduces OC differentiation. Using a c-Fos luciferase reporter assay lacking the TRE responsive element, we also demonstrate that M-CSF induces optimal c-Fos expression through DAG production. Finally, to demonstrate the importance of the M-CSF/DGKζ/DAG axis on regulation of c-Fos during osteoclastogenesis, we turned to PLCγ2(+/-) BMMs, which have reduced DAG levels and form fewer OCs because of impaired expression of the master regulator of osteoclastogenesis NFATc1 and c-Fos. Strikingly, genetic deletion of DGKζ in PLCγ2(+/-) mice rescues OC formation and normalizes c-Fos levels without altering NFATc1 expression. To our knowledge, this is the first report implicating M-CSF/DGKζ/DAG axis as a critical regulator of bone homeostasis via its actions on OC differentiation and c-Fos expression.
Collapse
Affiliation(s)
- Ali Zamani
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Corinne Decker
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Viviana Cremasco
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Lindsey Hughes
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Deborah V. Novack
- Department of Pathology and Immunology; Washington University School of Medicine; St. Louis, MO, 63110; USA
| | - Roberta Faccio
- Department of Orthopaedics; Washington University School of Medicine; St. Louis, MO, 63110; USA
- Corresponding Author Roberta Faccio, Box 8233, 660 S. Euclid, St. Louis, MO 63110, USA, Phone: 314-747-4602, Fax: 314-362-0334,
| |
Collapse
|
8
|
Lin J, Lee D, Choi Y, Lee SY. The scaffold protein RACK1 mediates the RANKL-dependent activation of p38 MAPK in osteoclast precursors. Sci Signal 2015; 8:ra54. [PMID: 26038599 DOI: 10.1126/scisignal.2005867] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
The E3 ubiquitin ligase TRAF6 [tumor necrosis factor (TNF) receptor (TNFR)-associated factor 6] and the associated kinase TAK1 [transforming growth factor-β (TGF-β)-activated kinase 1] are key components of the signaling pathways that activate nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPKs) in response to various stimuli. The cytokine RANKL (receptor activator of NF-κB ligand) is essential for the differentiation of bone marrow cells into bone-resorbing osteoclasts through the activation of NF-κB and MAPK. We found that the scaffold protein RACK1 (receptor for activated C kinase 1) selectively mediated the RANKL-dependent activation of p38 MAPK through the TRAF6-TAK1 axis by interacting with the MAPK kinase MKK6 (MAPK kinase kinase 6), which is upstream of p38 MAPK. RACK1 was necessary for the differentiation of bone marrow cells into osteoclasts through the stimulation of p38 MAPK activation. Osteoclast precursors exposed to RANKL exhibited an interaction among RACK1, RANK, TRAF6, TAK1, and the kinase MKK6, thereby leading to the activation of the MKK6-p38 MAPK pathway. Experiments in which RACK1 or TAK1 was knocked down in osteoclast precursors indicated that RACK1 acted as a bridge, bringing MKK6 to the TRAF6-TAK1 complex. Furthermore, local administration of RACK1-specific small interfering RNA (siRNA) into mice calvariae reduced the RANKL-induced bone loss by reducing the numbers of osteoclasts. These findings suggest that RACK1 specifies the RANKL-stimulated activation of p38 MAPK by facilitating the association of MKK6 with TAK1 and may provide a molecular target for a new therapeutic strategy to treat bone diseases.
Collapse
Affiliation(s)
- Jingjing Lin
- Department of Life Science, Ewha Womans University, Seoul 120-750, South Korea. Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, South Korea
| | - Daekee Lee
- Department of Life Science, Ewha Womans University, Seoul 120-750, South Korea
| | - Yongwon Choi
- Department of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | - Soo Young Lee
- Department of Life Science, Ewha Womans University, Seoul 120-750, South Korea. Research Center for Cellular Homeostasis, Ewha Womans University, Seoul 120-750, South Korea.
| |
Collapse
|
9
|
Yao J, Li J, Zhou L, Cheng J, Chim SM, Zhang G, Quinn JMW, Tickner J, Zhao J, Xu J. Protein kinase C inhibitor, GF109203X attenuates osteoclastogenesis, bone resorption and RANKL-induced NF-κB and NFAT activity. J Cell Physiol 2015; 230:1235-42. [PMID: 25363829 DOI: 10.1002/jcp.24858] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 10/28/2014] [Indexed: 12/14/2022]
Abstract
Osteolytic bone diseases are characterized by excessive osteoclast formation and activation. Protein kinase C (PKC)-dependent pathways regulate cell growth, differentiation and apoptosis in many cellular systems, and have been implicated in cancer development and osteoclast formation. A number of PKC inhibitors with anti-cancer properties have been developed, but whether they might also influence osteolysis (a common complication of bone invading cancers) is unclear. We studied the effects of the PKC inhibitor compound, GF109203X on osteoclast formation and activity, processes driven by receptor activator of NFκB ligand (RANKL). We found that GF109203X strongly and dose dependently suppresses osteoclastogenesis and osteoclast activity in RANKL-treated primary mouse bone marrow cells. Consistent with this GF109203X reduced expression of key osteoclastic genes, including cathepsin K, calcitonin receptor, tartrate resistant acid phosphatase (TRAP) and the proton pump subunit V-ATPase-d2 in RANKL-treated primary mouse bone marrow cells. Expression of these proteins is dependent upon RANKL-induced NF-κB and NFAT transcription factor actions; both were reduced in osteoclast progenitor populations by GF109203X treatment, notably NFATc1 levels. Furthermore, we showed that GF109203X inhibits RANKL-induced calcium oscillation. Together, this study shows GF109203X may block osteoclast functions, suggesting that pharmacological blockade of PKC-dependent pathways has therapeutic potential in osteolytic diseases.
Collapse
Affiliation(s)
- Jun Yao
- Department of Orthopaedic Surgery, Research Centre for Regenerative Medicine, The First Affiliated Hospital of Guangxi Medical University, Guangxi, China; School of Pathology and Laboratory Medicine, The University of Western Australia, Perth, Western Australia, Australia
| | | | | | | | | | | | | | | | | | | |
Collapse
|
10
|
Shin J, Jang H, Lin J, Lee SY. PKCβ positively regulates RANKL-induced osteoclastogenesis by inactivating GSK-3β. Mol Cells 2014; 37:747-52. [PMID: 25256217 PMCID: PMC4213766 DOI: 10.14348/molcells.2014.0220] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Revised: 08/18/2014] [Accepted: 08/18/2014] [Indexed: 12/14/2022] Open
Abstract
Protein kinase C (PKC) family members phosphorylate a wide variety of protein targets and are known to be involved in diverse cellular signaling pathways. However, the role of PKC in receptor activator of NF-κB ligand (RANKL) signaling has remained elusive. We now demonstrate that PKCβ acts as a positive regulator which inactivates glycogen synthase kinase-3β (GSK-3β) and promotes NFATc1 induction during RANKL-induced osteoclastogenesis. Among PKCs, PKCβ expression is increased by RANKL. Pharmacological inhibition of PKCβ decreased the formation of osteoclasts which was caused by the inhibition of NFATc1 induction. Importantly, the phosphorylation of GSK-3β was decreased by PKCβ inhibition. Likewise, down-regulation of PKCβ by RNA interference suppressed osteoclast differentiation, NFATc1 induction, and GSK-3β phosphorylation. The administration of PKC inhibitor to the RANKL-injected mouse calvaria efficiently protected RANKL-induced bone destruction. Thus, the PKCβ pathway, leading to GSK-3β inactivation and NFATc1 induction, has a key role in the differentiation of osteoclasts. Our results also provide a further rationale for PKCβ's therapeutic targeting to treat inflammation-related bone diseases.
Collapse
Affiliation(s)
- Jihye Shin
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
| | - Hyunduk Jang
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
- Present address: Department of Neurology, Seoul National University Hospital, and College of Medicine and Neuroscience Research Institute, Medical Research Center, Seoul National University, Seoul 110-749,
Korea
| | - Jingjing Lin
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
| | - Soo Young Lee
- Department of Life Science and the Research Center for Cellular Home-ostasis, Ewha Womans University, Seoul 120-750,
Korea
| |
Collapse
|
11
|
Khor EC, Abel T, Tickner J, Chim SM, Wang C, Cheng T, Ng B, Ng PY, Teguh DA, Kenny J, Yang X, Chen H, Nakayama KI, Nakayama K, Pavlos N, Zheng MH, Xu J. Loss of protein kinase C-δ protects against LPS-induced osteolysis owing to an intrinsic defect in osteoclastic bone resorption. PLoS One 2013; 8:e70815. [PMID: 23951014 PMCID: PMC3738588 DOI: 10.1371/journal.pone.0070815] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 06/24/2013] [Indexed: 11/18/2022] Open
Abstract
Bone remodeling is intrinsically regulated by cell signaling molecules. The Protein Kinase C (PKC) family of serine/threonine kinases is involved in multiple signaling pathways including cell proliferation, differentiation, apoptosis and osteoclast biology. However, the precise involvement of individual PKC isoforms in the regulation of osteoclast formation and bone homeostasis remains unclear. Here, we identify PKC-δ as the major PKC isoform expressed among all PKCs in osteoclasts; including classical PKCs (-α, -β and -γ), novel PKCs (-δ, -ε, -η and -θ) and atypical PKCs (-ι/λ and -ζ). Interestingly, pharmacological inhibition and genetic ablation of PKC-δ impairs osteoclastic bone resorption in vitro. Moreover, disruption of PKC-δ activity protects against LPS-induced osteolysis in mice, with osteoclasts accumulating on the bone surface failing to resorb bone. Treatment with the PKC-δ inhibitor Rottlerin, blocks LPS-induced bone resorption in mice. Consistently, PKC-δ deficient mice exhibit increased trabeculae bone containing residual cartilage matrix, indicative of an osteoclast-rich osteopetrosis phenotype. Cultured ex vivo osteoclasts derived from PKC-δ null mice exhibit decreased CTX-1 levels and MARKS phosphorylation, with enhanced formation rates. This is accompanied by elevated gene expression levels of cathepsin K and PKC -α, -γ and -ε, as well as altered signaling of pERK and pcSrc416/527 upon RANKL-induction, possibly to compensate for the defects in bone resorption. Collectively, our data indicate that PKC-δ is an intrinsic regulator of osteoclast formation and bone resorption and thus is a potential therapeutic target for pathological osteolysis.
Collapse
Affiliation(s)
- Ee Cheng Khor
- Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Tamara Abel
- Centre for Microscopy, Characterisation and Analysis, University of Western Australia, Nedlands, Western Australia, Australia
| | - Jennifer Tickner
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Shek Man Chim
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Cathy Wang
- Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
| | - Taksum Cheng
- Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
| | - Benjamin Ng
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Pei Ying Ng
- Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
| | - Dian Astari Teguh
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Jacob Kenny
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
| | - Xiaohong Yang
- Guangzhou Institute of Traumatic Surgery, the Fourth Affiliated Hospital of Medical College, Jinan University, Guangzhou, China
| | - Honghui Chen
- Guangzhou Institute of Traumatic Surgery, the Fourth Affiliated Hospital of Medical College, Jinan University, Guangzhou, China
| | - Keiichi I. Nakayama
- Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Keiko Nakayama
- Department of Molecular Genetics, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Nathan Pavlos
- Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
| | - Ming H. Zheng
- Centre for Orthopaedic Research, School of Surgery, University of Western Australia, Nedlands, Western Australia, Australia
- * E-mail: (JX); (MHZ)
| | - Jiake Xu
- School of Pathology and Laboratory Medicine, University of Western Australia, Nedlands, Western Australia, Australia
- * E-mail: (JX); (MHZ)
| |
Collapse
|
12
|
Lacombe J, Karsenty G, Ferron M. Regulation of lysosome biogenesis and functions in osteoclasts. Cell Cycle 2013; 12:2744-52. [PMID: 23966172 DOI: 10.4161/cc.25825] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In order to resorb the mineralized bone extracellular matrix, the osteoclast relies on the generation of a resorption lacuna characterized by the presence of specific proteases and a low pH. Hence, bone resorption by osteoclasts is highly dependent on lysosomes, the organelles specialized in intra- and extracellular material degradation. This is best illustrated by the fact that multiple forms of human osteopetrosis are caused by mutations in genes encoding for lysosomal proteins. Yet, until recently, the molecular mechanisms regulating lysosomal biogenesis and function in osteoclasts were poorly understood. Here we review the latest developments in the study of lysosomal biogenesis and function in osteoclasts with an emphasis on the transcriptional control of these processes.
Collapse
Affiliation(s)
- Julie Lacombe
- Institut de Recherches Cliniques de Montréal; Montréal, Québec, Canada
| | | | | |
Collapse
|
13
|
Ferron M, Settembre C, Shimazu J, Lacombe J, Kato S, Rawlings DJ, Ballabio A, Karsenty G. A RANKL-PKCβ-TFEB signaling cascade is necessary for lysosomal biogenesis in osteoclasts. Genes Dev 2013; 27:955-69. [PMID: 23599343 DOI: 10.1101/gad.213827.113] [Citation(s) in RCA: 146] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Bone resorption by osteoclasts requires a large number of lysosomes that release proteases in the resorption lacuna. Whether lysosomal biogenesis is a consequence of the action of transcriptional regulators of osteoclast differentiation or is under the control of a different and specific transcriptional pathway remains unknown. We show here, through cell-based assays and cell-specific gene deletion experiments in mice, that the osteoclast differentiation factor RANKL promotes lysosomal biogenesis once osteoclasts are differentiated through the selective activation of TFEB, a member of the MITF/TFE family of transcription factors. This occurs following PKCβ phosphorylation of TFEB on three serine residues located in its last 15 amino acids. This post-translational modification stabilizes and increases the activity of this transcription factor. Supporting these biochemical observations, mice lacking in osteoclasts--either TFEB or PKCβ--show decreased lysosomal gene expression and increased bone mass. Altogether, these results uncover a RANKL-dependent signaling pathway taking place in differentiated osteoclasts and culminating in the activation of TFEB to enhance lysosomal biogenesis-a necessary step for proper bone resorption.
Collapse
Affiliation(s)
- Mathieu Ferron
- Department of Genetics and Development, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
14
|
Kato K, Morita I. Acidosis environment promotes osteoclast formation by acting on the last phase of preosteoclast differentiation: a study to elucidate the action points of acidosis and search for putative target molecules. Eur J Pharmacol 2011; 663:27-39. [PMID: 21575626 DOI: 10.1016/j.ejphar.2011.04.062] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Revised: 04/14/2011] [Accepted: 04/18/2011] [Indexed: 01/06/2023]
Abstract
Acidosis promoted tartaric acid-resistant acid phosphatase-positive multinuclear cell (TRAP+MNC) or osteoclast formation. Large osteoclast or TRAP+LMNC formation was observed far more in an acidosis environment than in a physiologically neutral environment. One of the major action points of acidosis was determined to be located in the last phase of preosteoclast differentiation using a co-culture system and a soluble RANKL-dependent bone marrow cell culture system. On-going osteoclast formation in an acidosis environment markedly deteriorated when the medium was replaced with physiologically neutral medium within the first 6h; however, bone marrow cells previously stimulated in an acidosis environment for 9h differentiated into TRAP+LMNC in pH 7.4 medium. Messenger RNA (mRNA) expression levels of DC-STAMP, a key molecule in cell fusion, and NFATc1 did not increase in the acidosis environment compared with those under physiologically neutral conditions. Ruthenium red, a general TRP antagonist, deteriorated acidosis-promoted TRAP+LMNC formation. 4-Alpha-PDD, a TRPV4-specific agonist, added in the last 21 h of preosteoclast differentiation, potentiated TRAP+LMNC formation in a mild acidosis environment, showing synergism between TRPV4 activation and acidosis. RN1734, a TRPV4-specific antagonist, partly inhibited acidosis-promoted TRAP+LMNC formation. We thus narrowed down the major action points of acidosis in osteoclast formation and elucidated the characteristics of this system in detail. Our results show that acidosis effectively uses TRPV4 to drive large-scale cell fusion and also utilizes systems independently of TRPV4.
Collapse
Affiliation(s)
- Kohtaro Kato
- Department of Cellular Physiological Chemistry, Graduate School, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8549, Japan.
| | | |
Collapse
|
15
|
Fu Y, Zhao Y, Luan W, Dong LY, Dong Y, Lai B, Zhu Y, Zheng P. Sigma-1 receptors amplify dopamine D1 receptor signaling at presynaptic sites in the prelimbic cortex. BIOCHIMICA ET BIOPHYSICA ACTA 2010; 1803:1396-408. [PMID: 20732358 DOI: 10.1016/j.bbamcr.2010.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2010] [Revised: 07/28/2010] [Accepted: 08/16/2010] [Indexed: 12/01/2022]
Abstract
Sigma-1 receptors are highly expressed in the brain. The downstream signaling mechanisms associated with the sigma-1 receptor activation have been shown to involve the activation of protein kinase C (PKC), the control of Ca(2) homoeostasis and the regulation of voltage- and ligand-gated ion channels. But few studies examined the regulatory effect of sigma-1 receptors on metabotropic receptor signaling. The present paper studied the regulatory effect of sigma-1 receptors on the signaling of dopamine D1 receptors, one of metabotropic receptors, by examining the effect of sigma-1 receptor agonists on the D1 receptor agonist-induced cAMP-dependent protein kinase (PKA) activation at presynaptic sites using the synaptosomes from the prelimbic cortex. The results showed that sigma-1 receptor agonists alone had no effects on the PKA activity, but could amplify the D1 receptor agonist-induced PKA activation. The sigma-1 receptor agonist also amplified the membrane-permeable analog of cAMP- and the adenylyl cyclase (AC) activator-induced PKA activation, but did not on the D1 receptor agonist-induced AC activation. The conventional PKC (cPKC), especially the PKCβI, and the extracellular Ca(2+) influx through L-type Ca(2+) channels might play key roles in the amplifying effect of the sigma-1 receptor agonists. The activation of PKC by sigma-1 receptor agonists was the upstream event of the increase in the intrasynaptosomal Ca(2+) concentration. These results suggest that sigma-1 receptors may amplify the D1 receptor agonist-induced PKA activation by sigma-1 receptors - cPKC (especially the PKCβI) - L-type Ca(2+) channels - Ca(2+) - AC and/or cAMP signaling pathway.
Collapse
Affiliation(s)
- Yingmei Fu
- State key Laboratory of Medical Neurobiology, Shanghai Medical College and Institutes of Brain Science, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Kim MH, Park M, Baek SH, Kim HJ, Kim SH. Molecules and signaling pathways involved in the expression of OC-STAMP during osteoclastogenesis. Amino Acids 2010; 40:1447-59. [DOI: 10.1007/s00726-010-0755-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2010] [Accepted: 09/15/2010] [Indexed: 10/19/2022]
|
17
|
Wu PY, Lai B, Dong Y, Wang ZM, Li ZC, Zheng P. Different oxidants and PKC isozymes mediate the opposite effect of inhibition of Qi and Qo site of mitochondrial complex III on calcium currents in rat cortical neurons. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2010; 1803:1072-82. [DOI: 10.1016/j.bbamcr.2010.05.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2010] [Revised: 04/29/2010] [Accepted: 05/03/2010] [Indexed: 10/19/2022]
|
18
|
Hsu YL, Hung JY, Ko YC, Hung CH, Huang MS, Kuo PL. Phospholipase D signaling pathway is involved in lung cancer-derived IL-8 increased osteoclastogenesis. Carcinogenesis 2010; 31:587-96. [PMID: 20106902 DOI: 10.1093/carcin/bgq030] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Bone is a frequent target of lung cancer metastasis, which is associated with significant morbidity and a dismal prognosis. This study analyzed the soluble factors secreted by lung cancer cells, which are responsible for increasing osteoclast differentiation. Addition of recombinant human interleukin-8 (rhIL-8), present in large amounts in A549-conditioned medium (CM) and NCI-H460-CM, mimicked the inductive effect of A549-CM and NCI-H460-CM on osteoclastogenesis. In contrast, depletion of interleukin-8 (IL-8) from A549-CM and NCI-H460-CM decreased the osteoclastogenesis-inductive properties of A549-CM and NCI-H460-CM. Induction of osteoclast differentiation by lung cancer-derived-CM and rhIL-8 was associated with increased phospholipase D (PLD) activation, and the activations of protein kinase C (PKC) alpha/betaII, extracellular signal-regulated kinase (ERK) 1/2 and AKT/the mammalian target of rapamycin (mTOR). Blocking PLD by a specific inhibitor significantly decreased osteoclast formation by inhibiting PKCs activation and subsequently attenuating the phosphorylation of ERK1/2. PLD inhibitor also completely decreased AKT and mTOR phosphorylation, whereas phosphatidylinositol-3-kinase (PI3K) inhibitor only partially decreased mTOR phosphorylation, suggesting that mTOR activation by PLD is through both PI3K/AKT-dependent and PI3K/AKT-independent manner. In addition, blocking AKT and ERK1/2 by a specific inhibitor also suppressed lung cancer-derived-CM and rhIL-8-induced osteoclast differentiation. Moreover, treatment of peripheral blood mononuclear cells with sera from invasive lung cancer patients increased the formation of osteoclasts. Our study suggests that IL-8 or IL-8-mediated PLD/PKC/ERK1/2 or PLD/AKT signaling is an attractive therapeutic target for osteolytic bone metastases in lung cancer patients.
Collapse
Affiliation(s)
- Ya-Ling Hsu
- Graduate Institute of Medicine, Schoolof Medicine, College of Medicine, Kaohsiung Medical University, 807 Kaohsiung, Taiwan
| | | | | | | | | | | |
Collapse
|
19
|
Ang ESM, Pavlos NJ, Chai LY, Qi M, Cheng TS, Steer JH, Joyce DA, Zheng MH, Xu J. Caffeic acid phenethyl ester, an active component of honeybee propolis attenuates osteoclastogenesis and bone resorption via the suppression of RANKL-induced NF-kappaB and NFAT activity. J Cell Physiol 2009; 221:642-9. [PMID: 19681045 DOI: 10.1002/jcp.21898] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Receptor activator NF-kappaB ligand (RANKL)-activated signaling is essential for osteoclast differentiation, activation and survival. Caffeic acid phenethyl ester (CAPE), a natural NF-kappaB inhibitor from honeybee propolis has been shown to have anti-tumor and anti-inflammatory properties. In this study, we investigated the effect of CAPE on the regulation of RANKL-induced osteoclastogenesis, bone resorption and signaling pathways. Low concentrations of CAPE (<1 microM) dose dependently inhibited RANKL-induced osteoclastogenesis in RAW264.7 cell and bone marrow macrophage (BMM) cultures, as well as decreasing the capacity of human osteoclasts to resorb bone. CAPE inhibited both constitutive and RANKL-induced NF-kappaB and NFAT activation, concomitant with delayed IkappaBalpha degradation and inhibition of p65 nuclear translocation. At higher concentrations, CAPE induced apoptosis and caspase 3 activities of RAW264.7 and disrupts the microtubule network in osteoclast like (OCL) cells. Taken together, our findings demonstrate that inhibition of NF-kappaB and NFAT activation by CAPE results in the attenuation of osteoclastogenesis and bone resorption, implying that CAPE is a potential treatment for osteolytic bone diseases.
Collapse
Affiliation(s)
- Estabelle S M Ang
- Molecular Orthopaedic Laboratory, Centre for Orthopaedic Research, School of Surgery, The University of Western Australia, Nedlands, Western Australia, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Tiedemann K, Hussein O, Sadvakassova G, Guo Y, Siegel PM, Komarova SV. Breast cancer-derived factors stimulate osteoclastogenesis through the Ca2+/protein kinase C and transforming growth factor-beta/MAPK signaling pathways. J Biol Chem 2009; 284:33662-70. [PMID: 19801662 DOI: 10.1074/jbc.m109.010785] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Breast cancer commonly metastasizes to bone where its growth depends on the action of bone-resorbing osteoclasts. We have previously shown that breast cancer cells secrete factors able to directly stimulate osteoclastogenesis from receptor activator of nuclear factor kappaB ligand (RANKL)-primed precursors and that transforming growth factor-beta (TGFbeta) plays a permissive role in this process. Now, we evaluate the signaling events triggered in osteoclast precursors by soluble factors produced by MDA-MB-231 human breast carcinoma cells. In mouse bone marrow cultures and RAW 264.7 murine monocytic cells, MDA-MB-231-derived factors increased osteoclast number, size, and nucleation. These factors failed to induce Smad2 phosphorylation, and short interfering RNAs against Smad4 did not affect their ability to induce osteoclastogenesis. In contrast, MDA-MB-231 factors induced phosphorylation of p38 and ERK1/2, and pharmacological inhibitors against p38 (SB203580) and MEK1/2 (PD98059) impeded the osteoclastogenic effects of cancer-derived factors. Neutralizing antibodies against TGFbeta attenuated p38 activation, whereas activation of ERK1/2 was shortened in duration, but not decreased in amplitude. ERK1/2 phosphorylation induced by cancer-derived factors was blocked by MEK1/2 inhibitor, but not by Ras (manumycin A) or Raf (GW5074) inhibitors. Inhibition of protein kinase Calpha using Gö6976 prevented both ERK1/2 phosphorylation and osteoclast formation in response to MDA-MB-231-derived factors. Using microspectrofluorimetry of fura-2-AM-loaded osteoclast precursors, we have found that cancer-derived factors, similar to RANKL, induced sustained oscillations in cytosolic free calcium. The calcium chelator BAPTA prevented calcium elevations and osteoclast formation in response to MDA-MB-231-derived factors. Thus, we have shown that breast cancer-derived factors induce osteoclastogenesis through the activation of calcium/protein kinase Calpha and TGFbeta-dependent ERK1/2 and p38 signaling pathways.
Collapse
Affiliation(s)
- Kerstin Tiedemann
- Faculty of Dentistry, McGill University, Montreal, Quebec H3A 2B2, Canada
| | | | | | | | | | | |
Collapse
|
21
|
Hurtel-Lemaire AS, Mentaverri R, Caudrillier A, Cournarie F, Wattel A, Kamel S, Terwilliger EF, Brown EM, Brazier M. The Calcium-sensing Receptor Is Involved in Strontium Ranelate-induced Osteoclast Apoptosis. J Biol Chem 2009; 284:575-584. [DOI: 10.1074/jbc.m801668200] [Citation(s) in RCA: 185] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
22
|
Armstrong S, Pereverzev A, Dixon SJ, Sims SM. Activation of P2X7 receptors causes isoform-specific translocation of protein kinase C in osteoclasts. J Cell Sci 2008; 122:136-44. [PMID: 19066285 DOI: 10.1242/jcs.031534] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Nucleotides, released in response to mechanical or inflammatory stimuli, signal through P2 nucleotide receptors in many cell types. Osteoclasts express P2X7 receptors (encoded by P2rx7) - Ca(2+)-permeable channels that are activated by high concentrations of extracellular ATP. Genetic disruption of P2rx7 leads to increased resorption and reduced skeletal response to mechanical stimuli. To investigate whether P2X7 receptors couple to activation of protein kinase C (PKC), RAW 264.7 cells were differentiated into multinucleated osteoclast-like cells and live-cell confocal imaging was used to localize enhanced green fluorescent protein (EGFP)-tagged PKC. Benzoylbenzoyl-ATP (BzATP; a P2X7 agonist) induced transient translocation of PKCalpha to the basolateral membrane. UTP or ATP (10 microM), which activate P2 receptors other than P2X7, failed to induce translocation. Moreover, BzATP failed to induce PKC translocation in osteoclasts derived from the bone marrow of P2rx7(-/-) mice, demonstrating specificity for P2X7. BzATP induced a transient rise of cytosolic Ca(2+), and removal of extracellular Ca(2+) abolished the translocation of PKCalpha that was induced by BzATP (but not by phorbol ester). We examined the isoform specificity of this response, and observed translocation of the Ca(2+)-dependent isoforms PKCalpha and PKCbetaI, but not the Ca(2+)-independent isoform PKCdelta. Thus, activation of P2X7 receptors specifically induces Ca(2+)-dependent translocation of PKC to the basolateral membrane domain of osteoclasts, an aspect of spatiotemporal signaling not previously recognized.
Collapse
Affiliation(s)
- Souzan Armstrong
- Department of Physiology and Pharmacology, Schulich School of Medicine and Dentistry, The University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | | | |
Collapse
|
23
|
Kamon M, Fujita D, Goto N, Amano H, Sakamoto K. Prostaglandin F2α negatively regulates bone resorption in murine osteoclast development. Prostaglandins Other Lipid Mediat 2008; 87:26-33. [DOI: 10.1016/j.prostaglandins.2008.06.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2008] [Revised: 03/25/2008] [Accepted: 06/12/2008] [Indexed: 10/21/2022]
|
24
|
Pereverzev A, Komarova SV, Korcok J, Armstrong S, Tremblay GB, Dixon SJ, Sims SM. Extracellular acidification enhances osteoclast survival through an NFAT-independent, protein kinase C-dependent pathway. Bone 2008; 42:150-61. [PMID: 17964236 DOI: 10.1016/j.bone.2007.08.044] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2007] [Revised: 07/24/2007] [Accepted: 08/31/2007] [Indexed: 11/22/2022]
Abstract
Systemic acidosis has detrimental effects on the skeleton and local acidosis is associated with bone destruction in inflammatory and neoplastic diseases. However, the mechanisms by which acidosis enhances osteoclastic bone resorption are poorly understood. Our aim was to examine the effects of acid on osteoclast survival and the involvement of cytosolic Ca(2+) in mediating these effects. Osteoclasts were isolated from long bones of newborn rats, and multinucleated osteoclast-like cells were generated from RAW 264.7 cells. Cytosolic free Ca(2+) concentration ([Ca(2+)](i)) was monitored using fura-2. Survival of rat osteoclasts over a period of 18 h was significantly enhanced by acidification of the medium from 40+/-10% at pH 7.6 to 83+/-4% at pH 7.0. Consistent with its effects on survival, acidosis suppressed osteoclast apoptosis at 6 h. We examined the possible involvement of the proton-sensing receptor ovarian cancer G protein-coupled receptor 1 (OGR1) in mediating the effects of acid. Acid-induced rise of [Ca(2+)](i) was inhibited by the OGR1 antagonist Cu(2+) and was suppressed in osteoclast-like cells in which OGR1 transcripts were depleted using RNA interference. These findings support an essential role for OGR1 in acid-induced Ca(2+) signaling in osteoclasts. Addition of Cu(2+) or chelation of cytosolic Ca(2+) with BAPTA abolished the ability of acidification to enhance osteoclast survival. Inhibition of NFAT activation with the cell-permeable peptide 11R-VIVIT did not alter the ability of acid to promote survival; however, it suppressed the increase in survival induced by RANKL. In contrast, inhibition of protein kinase C (PKC) blocked the effect of acid on osteoclast survival. Thus, this study reveals that extracellular acidification enhances osteoclast survival through an NFAT-independent, PKC-dependent pathway. Increased osteoclast survival may contribute to bone loss in systemic and local acidosis.
Collapse
Affiliation(s)
- Alexey Pereverzev
- CIHR Group in Skeletal Development and Remodeling, Schulich School of Medicine & Dentistry, The University of Western Ontario, London, Ontario, Canada N6A 5C1
| | | | | | | | | | | | | |
Collapse
|
25
|
Dudek AZ, Zwolak P, Jasinski P, Terai K, Gallus NJ, Ericson ME, Farassati F. Protein kinase C-β inhibitor enzastaurin (LY317615.HCI) enhances radiation control of murine breast cancer in an orthotopic model of bone metastasis. Invest New Drugs 2007; 26:13-24. [PMID: 17805485 DOI: 10.1007/s10637-007-9079-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2007] [Accepted: 08/06/2007] [Indexed: 10/22/2022]
Abstract
Radiation therapy is a widely used treatment for metastatic bone cancer, but the rapid onset of tumor radioresistance is a major problem. We investigated the radiosensitizing effect of enzastaurin, a protein kinase Cbeta (PKCbeta) inhibitor, on bone tumor growth and tumor-related pain. We found that enzastaurin enhanced the effect of ionizing radiation on cultured murine 4T1 breast cancer and murine endothelial cells, suppressing their proliferation and colony formation. Enzastaurin and ionizing radiation also induced caspase-mediated apoptosis of 4T1 cells to a greater degree than radiation alone. Enzastaurin treatment of 4T1 cells blocked the phosphorylation of PKCbeta, as well as Ras and two of its downstream effectors ERK1/2 and RAL-GTP. Using an orthotopic model of bone metastasis, we observed that a combination of enzastaurin and localized radiation treatment reduced tumor blood vessel density, bone destruction and pain compared to single modality treatment. In conclusion, we demonstrate that inhibition of PKCbeta in combination with localized radiation treatment suppresses tumor growth and alleviates pain as compared to radiation-only treatment. We also show that the radiosensitizing effect of enzastaurin is associated with suppression of tumor cell proliferation and tumor-induced angiogenesis possibly through inhibition of the Ras pathway.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/radiation effects
- Bone Neoplasms/drug therapy
- Bone Neoplasms/radiotherapy
- Bone Neoplasms/secondary
- Caspases/metabolism
- Cell Line, Tumor
- Cell Survival/drug effects
- Cell Survival/radiation effects
- Combined Modality Therapy
- Disease Models, Animal
- Dose-Response Relationship, Drug
- Endothelial Cells/drug effects
- Endothelial Cells/radiation effects
- Female
- Indoles/pharmacology
- Indoles/therapeutic use
- Mammary Neoplasms, Experimental/drug therapy
- Mammary Neoplasms, Experimental/pathology
- Mammary Neoplasms, Experimental/radiotherapy
- Mice
- Mice, Inbred C3H
- Mice, SCID
- Neovascularization, Pathologic/drug therapy
- Neovascularization, Pathologic/radiotherapy
- Osteoclasts/drug effects
- Osteoclasts/metabolism
- Osteoclasts/radiation effects
- Pain/drug therapy
- Pain/radiotherapy
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C beta
- Radiation-Sensitizing Agents/pharmacology
- Radiation-Sensitizing Agents/therapeutic use
- Radiotherapy, Adjuvant/methods
- Signal Transduction/drug effects
- Tumor Burden/drug effects
- Tumor Burden/radiation effects
- ras Proteins/metabolism
Collapse
Affiliation(s)
- Arkadiusz Z Dudek
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota, Mayo Mail Code 480, 420 Delaware Street SE, Minneapolis, MN 55455, USA.
| | | | | | | | | | | | | |
Collapse
|
26
|
Majumder N, Dey R, Mathur RK, Datta S, Maitra M, Ghosh S, Saha B, Majumdar S. An unusual pro-inflammatory role of interleukin-10 induced by arabinosylated lipoarabinomannan in murine peritoneal macrophages. Glycoconj J 2007; 23:675-86. [PMID: 17115274 DOI: 10.1007/s10719-006-9017-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
Various species of Mycobacteria produce a major cell wall-associated lipoglycan, called Lipoarabinomannan (LAM), which is involved in the virulence of Mycobacterial species. In this study, we tried to establish the role of the increased IL-10 secretion under Arabinosylated-LAM (Ara-LAM) treatment, the LAM that induces apoptosis in host macrophages or PBMC. We have studied the survival and apoptotic factors by western blotting, and estimated nitrite generation by Griess reaction, quantified iNOS mRNA by semi-quantitative RT-PCR, and ultimately the fate of the cells were studied by Flow Cytometric Analysis of AnnexinV-FITC binding. As per our observations, neutralization of released IL-10 in C57BL/6 peritoneal macrophages prior to Ara-LAM treatment, as well as macrophages from IL-10 knockout (KO) mice treated with Ara-LAM, showed significant down regulation of pro-apoptotic factors and up regulation of survival factors. These effects were strikingly similar to those when peritoneal macrophages were subjected to TNF-alpha and IL-12 neutralization followed by Ara-LAM-treatment. However, under similar conditions virulent Mannosylated-LAM (from Mycobacterium tuberculosis) treatment of macrophages clearly depicts the importance of IL-10 in the maintenance of pathogenesis, proving its usual immunosuppressive role. Thus, from our detailed investigations we point out an unusual pro-inflammatory action of IL-10 in Ara-LAM treated macrophages, where it behaves in a similar manner as the known Th1 cytokines TNF- alpha and IL-12.
Collapse
Affiliation(s)
- Nivedita Majumder
- Department of Microbiology, Bose Institute, P1/12 CIT Scheme VII M, Kolkata, India
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Chang EJ, Kim HJ, Ha J, Kim HJ, Ryu J, Park KH, Kim UH, Lee ZH, Kim HM, Fisher DE, Kim HH. Hyaluronan inhibits osteoclast differentiation via Toll-like receptor 4. J Cell Sci 2007; 120:166-76. [PMID: 17164294 DOI: 10.1242/jcs.03310] [Citation(s) in RCA: 86] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The differentiation of osteoclasts, cells specialized for bone resorption, is governed by two key factors, macrophage colony stimulating factor (M-CSF) and receptor activator of nuclear factor κB ligand (RANKL). The extracellular matrix (ECM) is an important factor influencing cell fate. To date, little investigation on the relationship between ECM components and osteoclast differentiation has been documented. In this study, we uncovered a potent anti-osteoclastogenic effect of hyaluronan (HA), an ECM component present in bone marrow and soft connective tissues, in primary mouse and human osteoclast precursor cell cultures. The anti-osteoclastogenic function of HA was dependent on Toll-like receptor 4 (TLR4) but not on CD44. HA inhibited M-CSF-dependent signaling pathways involving Rac, reactive oxygen species and mitogen-activated protein kinases, resulting in suppression of transcription factors AP-1 and MITF that control RANK expression. Furthermore, in an in vivo mouse model of calvarial bone resorption assays HA reduced RANKL-induced bone erosion and osteoclastogenesis. Our results clearly show that HA inhibits osteoclast differentiation through TLR4 by interfering with M-CSF signaling, and point that the interaction between ECM components and innate immune receptors can play an important role in the regulation of bone metabolism.
Collapse
Affiliation(s)
- Eun-Ju Chang
- Department of Cell and Developmental Biology and Brain Korea 21 Program, DRI, Seoul National University, Seoul 110-749, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Kim YH, Jun JH, Woo KM, Ryoo HM, Kim GS, Baek JH. Dexamethasone inhibits the formation of multinucleated osteoclastsvia down-regulation of β3 integrin expression. Arch Pharm Res 2006; 29:691-8. [PMID: 16964765 DOI: 10.1007/bf02968254] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Although glucocorticoids are known to affect osteoclast differentiation and function, there have been conflicting reports about the effect of glucocorticoids on osteoclast formation, leading to the assumption that microenvironment and cell type influence their action. We explored the effect of the synthetic glucocorticoid analog dexamethasone on the formation of osteoclasts. Dexamethasone inhibited the formation of tartrate-resistant acid phosphatase (TRAP)-positive multinucleated osteoclasts without affecting the formation of TRAP-positive mononuclear cells in a coculture of mouse osteoblasts and bone marrow cells. Dexamethasone did not inhibit mRNA expression levels of the receptor activator of nuclear factor-kappaB ligand and osteoprotegerin, the essential regulators of osteoclastogenesis. Dexamethasone down-regulated the expression of beta3 integrin mRNA and protein but did not alter expression of other osteoclast differentiation marker genes. Both dexamethasone and echistatin, a beta3 integrin function blocker, inhibited TRAP-positive multinucleated osteoclast formation but not TRAP-positive mononuclear cell formation. These results suggest that dexamethasone inhibits the formation of multinucleated osteoclasts, at least in part, through the down-regulation of beta3 integrin, which plays an important role in the formation of multinucleated osteoclasts.
Collapse
Affiliation(s)
- Yong Hee Kim
- Department of Cell and Developmental Biology, School of Dentistry and Dental Research Institute, Seoul National University, Seoul 110-749, Korea
| | | | | | | | | | | |
Collapse
|
29
|
Mookerjee Basu J, Mookerjee A, Sen P, Bhaumik S, Sen P, Banerjee S, Naskar K, Choudhuri SK, Saha B, Raha S, Roy S. Sodium antimony gluconate induces generation of reactive oxygen species and nitric oxide via phosphoinositide 3-kinase and mitogen-activated protein kinase activation in Leishmania donovani-infected macrophages. Antimicrob Agents Chemother 2006; 50:1788-97. [PMID: 16641451 PMCID: PMC1472228 DOI: 10.1128/aac.50.5.1788-1797.2006] [Citation(s) in RCA: 140] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Pentavalent antimony complexes, such as sodium stibogluconate and sodium antimony gluconate (SAG), are still the first choice for chemotherapy against various forms of leishmaniasis, including visceral leishmaniasis, or kala-azar. Although the requirement of a somewhat functional immune system for the antileishmanial action of antimony was reported previously, the cellular and molecular mechanism of action of SAG was not clear. Herein, we show that SAG induces extracellular signal-regulated kinase 1 (ERK-1) and ERK-2 phosphorylation through phosphoinositide 3-kinase (PI3K), protein kinase C, and Ras activation and p38 mitogen-activated protein kinase (MAPK) phosphorylation through PI3K and Akt activation. ERK-1 and ERK-2 activation results in an increase in the production of reactive oxygen species (ROS) 3 to 6 h after SAG treatment, while p38 MAPK activation and subsequent tumor necrosis factor alpha release result in the production of nitric oxide (NO) 24 h after SAG treatment. Thus, this study has provided the first evidence that SAG treatment induces activation of some important components of the intracellular signaling pathway, which results in an early wave of ROS-dependent parasite killing and a stronger late wave of NO-dependent parasite killing. This opens up the possibility of this metalloid chelate being used in the treatment of various diseases either alone or in combination with other drugs and vaccines.
Collapse
Affiliation(s)
- Jayati Mookerjee Basu
- Department of Immunology, Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Kolkata 700032, India.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Kim HJ, Chang EJ, Kim HM, Lee SB, Kim HD, Su Kim G, Kim HH. Antioxidant alpha-lipoic acid inhibits osteoclast differentiation by reducing nuclear factor-kappaB DNA binding and prevents in vivo bone resorption induced by receptor activator of nuclear factor-kappaB ligand and tumor necrosis factor-alpha. Free Radic Biol Med 2006; 40:1483-93. [PMID: 16632109 DOI: 10.1016/j.freeradbiomed.2005.10.066] [Citation(s) in RCA: 107] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2005] [Revised: 10/03/2005] [Accepted: 10/26/2005] [Indexed: 10/25/2022]
Abstract
The relationship between oxidative stress and bone mineral density or osteoporosis has recently been reported. As bone loss occurring in osteoporosis and inflammatory diseases is primarily due to increases in osteoclast number, reactive oxygen species (ROS) may be relevant to osteoclast differentiation, which requires receptor activator of nuclear factor-kappaB ligand (RANKL). Tumor necrosis factor-alpha (TNF-alpha) frequently present in inflammatory conditions has a profound synergy with RANKL in osteoclastogenesis. In this study, we investigated the effects of alpha-lipoic acid (alpha-LA), a strong antioxidant clinically used for some time, on osteoclast differentiation and bone resorption. At concentrations showing no growth inhibition, alpha-LA potently suppressed osteoclastogenesis from bone marrow-derived precursor cells driven either by a high-dose RANKL alone or by a low-dose RANKL plus TNF-alpha (RANKL/TNF-alpha). alpha-LA abolished ROS elevation by RANKL or RANKL/TNF-alpha and inhibited NF-kappaB activation in osteoclast precursor cells. Specifically, alpha-LA reduced DNA binding of NF-kappaB but did not inhibit IKK activation. Furthermore, alpha-LA greatly suppressed in vivo bone loss induced by RANKL or TNF-alpha in a calvarial remodeling model. Therefore, our data provide evidence that ROS plays an important role in osteoclast differentiation through NF-kappaB regulation and the antioxidant alpha-lipoic acid has a therapeutic potential for bone erosive diseases.
Collapse
Affiliation(s)
- Hyon Jong Kim
- Department of Cell and Developmental Biology, Brain Korea 21 Program, and Dental Research Institute, Seoul National University School of Dentistry, 28 Yeongon-Dong, Chongno-Gu, Seoul 110-749, Korea
| | | | | | | | | | | | | |
Collapse
|
31
|
García-Pérez MA, Noguera I, Hermenegildo C, Martínez-Romero A, Tarín JJ, Cano A. Alterations in the phenotype and function of immune cells in ovariectomy-induced osteopenic mice. Hum Reprod 2006; 21:880-7. [PMID: 16459351 DOI: 10.1093/humrep/dei413] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Within the last few years, much evidence has been presented on the involvement of the immune system in certain types of bone loss, such as activated T cells in rheumatoid arthritis and in periodontitis. Estrogen deficiency induces bone loss; however, how this deficiency affects the immune system has not been sufficiently studied. METHODS To evaluate the effects of estrogen withdrawal on the status and functionality of the immune system, mice were ovariectomized or sham-operated, and 5 weeks after surgery, when osteopenia had developed, several parameters were analysed in spleen and in bone marrow. We analysed bone turnover, cell phenotype by flow cytometry, cell function by cell proliferation assays, and the expression of several genes related to the process. RESULTS Five weeks after ovariectomy, augmented osteoclastogenesis persisted in the bone marrow. In addition, the ovariectomized mice had more B-cells and CD3+ T-cells expressing the receptor activator of NF-kappaB ligand (CD3+/RANKL+). The ovariectomized mice had lower serum alkaline phosphatase activity, a normal amount of T cells, lower percentages of CD11b+ and CD51+ cells in the bone marrow, and a lower serum interferon-gamma level compared with sham-operated controls. CONCLUSIONS The data suggest that, 5 weeks after ovariectomy, bone turnover remains imbalanced, with increased osteoclastogenesis and a decreased rate of bone formation. Moreover, there is an increase in B-cell formation, with normal and decreased percentages of T cells and myelomonocytic cells (CD11b+), respectively, in the bone marrow. Decreased serum interferon-gamma levels could be involved in the increased osteoclastogenesis found in the present work.
Collapse
Affiliation(s)
- M A García-Pérez
- Research Unit, Hospital Clínico Universitario of Valencia, Av. Blasco Ibáñez 17, 46010 Valencia, Spain.
| | | | | | | | | | | |
Collapse
|
32
|
Dey R, Sarkar A, Majumder N, Bhattacharyya Majumdar S, Roychoudhury K, Bhattacharyya S, Roy S, Majumdar S. Regulation of impaired protein kinase C signaling by chemokines in murine macrophages during visceral leishmaniasis. Infect Immun 2006; 73:8334-44. [PMID: 16299331 PMCID: PMC1307035 DOI: 10.1128/iai.73.12.8334-8344.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The protein kinase C (PKC) family regulates macrophage function involved in host defense against infection. In the case of Leishmania donovani infection, the impairment of PKC-mediated signaling is one of the crucial events for the establishment of parasite into the macrophages. Earlier reports established that C-C chemokines mediated protection against leishmaniasis via the generation of nitric oxide after 48 h. In this study, we investigated the role of MIP-1alpha and MCP-1 in the regulation of impaired PKC activity in the early hours (6 h) of infection. These chemokines restored Ca2+-dependent PKC activity and inhibited Ca2+-independent atypical PKC activity in L. donovani-infected macrophages under both in vivo and in vitro conditions. Pretreatment of macrophages with chemokines induced superoxide anion generation by activating NADPH oxidase components in infected cells. Chemokine administration in vitro induced the migration of infected macrophages and triggered the production of reactive oxygen species. In vivo treatment with chemokines significantly restricted the parasitic burden in livers as well as in spleens. Collectively, these results indicate a novel regulatory role of C-C chemokines in controlling the intracellular growth and multiplication of L. donovani, thereby demonstrating the antileishmanial properties of C-C chemokines in the disease process.
Collapse
Affiliation(s)
- Ranadhir Dey
- Department of Microbiology, Bose Institute, P1/12, C.I.T. Scheme VII-M, Kolkata-700 054, India
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Slattery C, Campbell E, McMorrow T, Ryan MP. Cyclosporine A-induced renal fibrosis: a role for epithelial-mesenchymal transition. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:395-407. [PMID: 16049326 PMCID: PMC1603578 DOI: 10.1016/s0002-9440(10)62984-7] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Cyclosporine A, which has been the foremost immunosuppressive agent since the early 1980's, significantly improves the success of organ transplantation. However, common complications of cyclosporine A therapy, such as severe renal tubulointerstitial fibrosis, limit the drug's clinical use. Although the exact mechanisms driving cyclosporine A-induced tubulointerstitial fibrosis remain elusive, we hypothesized that epithelial-mesenchymal transition (EMT) may play a major role. We investigated this in vitro by treating human proximal tubular cells with cyclosporine A. Morphological changes were observed after cyclosporine A treatment, including cell elongation (with a large degree of detachment), cytoskeletal rearrangement, and junctional disruption. In addition, expression of the myofibroblast-specific marker alpha-smooth muscle actin was detected in treated cells. These observations are consistent with events described during EMT. Using Affymetrix gene microarrays, we identified 128 genes that were differentially regulated in renal tubular cells after cyclosporine A treatment, including known profibrotic factors, oncogenes, and transcriptional regulators. Cyclosporine A induced a dose-dependent increase in transforming growth factor-beta secretion from proximal tubular cells. Subsequent functional studies revealed that protein kinase C-beta isoforms play a key role in cyclosporine A-induced effects. These findings provide novel insights into cyclosporine A-induced renal fibrosis and the molecular mechanisms underlying EMT, events that may be relevant in other disease states.
Collapse
Affiliation(s)
- Craig Slattery
- Department of Pharmacology, Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | |
Collapse
|
34
|
Kwan Tat S, Padrines M, Théoleyre S, Heymann D, Fortun Y. IL-6, RANKL, TNF-alpha/IL-1: interrelations in bone resorption pathophysiology. Cytokine Growth Factor Rev 2004; 15:49-60. [PMID: 14746813 DOI: 10.1016/j.cytogfr.2003.10.005] [Citation(s) in RCA: 698] [Impact Index Per Article: 33.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
All osteogenic cells (osteoclasts, osteoblasts) contribute individually to bone remodeling. Their cellular interactions control their cellular activities and the bone remodeling intensity. These interactions can be established either through a cell-cell contact, involving molecules of the integrin family, or by the release of many polypeptidic factors and/or their soluble receptor chains. These factors can act directly on osteogenic cells and their precursors to control differentiation, formation and functions (matrix formation, mineralization, resorption...). Here, we present the involvement of three groups of cytokines which seem to be of particular importance in bone physiology: interleukin-6 (IL-6), tumor necrosis factor-alpha (TNF-alpha) (TNF-alpha)/IL-1, and the more recently known triad osteoprotegerin (OPG)/receptor activator of NF-kappaB (RANK)/RANK ligand (RANKL). The interactions between these three groups are presented within the framework of bone resorption pathophysiology such as tumor associated osteolysis. The central role of the OPG/RANK/RANKL triad is pointed out.
Collapse
Affiliation(s)
- Steeve Kwan Tat
- Laboratoire de Physiopathologie de la Résorption Osseuse et Thérapie des Tumeurs Osseuses Primitives, Faculté de Médecine, 1 rue Gaston Veil, BP 53508, 44035 Nantes 1, France
| | | | | | | | | |
Collapse
|
35
|
Uchiyama S, Yamaguchi M. Inhibitory effect of β-cryptoxanthin on osteoclast-like cell formation in mouse marrow cultures. Biochem Pharmacol 2004; 67:1297-305. [PMID: 15013845 DOI: 10.1016/j.bcp.2003.11.011] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2003] [Accepted: 11/14/2003] [Indexed: 11/18/2022]
Abstract
The carotenoid beta-cryptoxanthin has been shown to have an inhibitory effect on bone-resorping factor-stimulated bone resorption in rat bone tissues in vitro. The effect of beta-cryptoxanthin on osteoclast-like cell formation in mouse marrow culture in vitro was investigated. The bone marrow cells were cultured for 7 days in alpha-minimal essential medium containing a bone-resorbing agent [parathyroid hormone (1-34) (PTH), prostaglandin E(2), 1,25-dihydroxyvitamin D(3), lipopolysaccharide, or tumor necrosis factor-alpha (TNFalpha)] with an effective concentration. Osteoclast-like cell formation was estimated by staining for tartrate-resistant acid phosphatase, a marker enzyme of osteoclasts. The presence of PTH (10(-7)M), prostaglandin E(2) (10(-5)M), 1,25-dihydroxyvitamin D(3) (10(-7)M), lipopolysaccharide (10 microg/mL), or TNFalpha (10 ng/mL) induced a remarkable increase in osteoclast-like multinucleated cells. These increases were significantly inhibited in the presence of beta-cryptoxanthin (10(-8) to 10(-6)M). beta-Cryptoxanthin (10(-7) and 10(-6)M) significantly inhibited dibutyryl cyclic adenosine monophosphate (DcAMP) (10(-5)M) or phorbol 12-myristate 13-acetate (PMA) (10(-5)M), an activator of protein kinase C, induced osteoclast-like cell formation. Also, beta-cryptoxanthin (10(-7) and 10(-6)M) had a significant inhibitory effect on osteoclast-like formation induced by receptor activator of NF-kappaB ligand (RANKL) (10 and 20 ng/mL) in the presence of macrophage colony-stimulating factor (M-CSF) (10 and 20 ng/mL). The stimulatory effect of RANKL and M-CSF on osteoclast-like cell formation was significantly enhanced in the presence of PMA, while such an effect was not seen by DcAMP. beta-Cryptoxanthin (10(-6)M) significantly inhibited osteoclast-like cell formation induced by RANKL and M-CSF in the presence of PMA or DcAMP. Moreover, the inhibitory effect of beta-cryptoxanthin on RANKL plus M-CSF-, PTH-, or TNFalpha-induced osteoclast-like cell formation was not observed in the presence of cycloheximide (10(-7)M), an inhibitor of protein synthesis at translational process, or 5,6-dichloro-1-beta-d-ribofuranosylbenzimidazole (10(-6)M), an inhibitor of transcription. This study demonstrates that beta-cryptoxanthin has a potent inhibitory effect on osteoclast-like cell formation in mouse marrow culture. The inhibitory action of beta-cryptoxanthin may partly involve in a newly synthesized protein component which is related to RANKL stimulation in osteoclastogenesis.
Collapse
Affiliation(s)
- Satoshi Uchiyama
- Laboratory of Endocrinology and Molecular Metabolism, Graduate School of Nutritional Sciences, University of Shizuoka, 52-1 Yada, Shizuoka 422-8526, Japan
| | | |
Collapse
|