1
|
Celik B, Leal AF, Tomatsu S. Potential Targeting Mechanisms for Bone-Directed Therapies. Int J Mol Sci 2024; 25:8339. [PMID: 39125906 PMCID: PMC11312506 DOI: 10.3390/ijms25158339] [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: 05/21/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/12/2024] Open
Abstract
Bone development is characterized by complex regulation mechanisms, including signal transduction and transcription factor-related pathways, glycobiological processes, cellular interactions, transportation mechanisms, and, importantly, chemical formation resulting from hydroxyapatite. Any abnormal regulation in the bone development processes causes skeletal system-related problems. To some extent, the avascularity of cartilage and bone makes drug delivery more challenging than that of soft tissues. Recent studies have implemented many novel bone-targeting approaches to overcome drawbacks. However, none of these strategies fully corrects skeletal dysfunction, particularly in growth plate-related ones. Although direct recombinant enzymes (e.g., Vimizim for Morquio, Cerezyme for Gaucher, Elaprase for Hunter, Mepsevii for Sly diseases) or hormone infusions (estrogen for osteoporosis and osteoarthritis), traditional gene delivery (e.g., direct infusion of viral or non-viral vectors with no modifications on capsid, envelope, or nanoparticles), and cell therapy strategies (healthy bone marrow or hematopoietic stem cell transplantation) partially improve bone lesions, novel delivery methods must be addressed regarding target specificity, less immunogenicity, and duration in circulation. In addition to improvements in bone delivery, potential regulation of bone development mechanisms involving receptor-regulated pathways has also been utilized. Targeted drug delivery using organic and inorganic compounds is a promising approach in mostly preclinical settings and future clinical translation. This review comprehensively summarizes the current bone-targeting strategies based on bone structure and remodeling concepts while emphasizing potential approaches for future bone-targeting systems.
Collapse
Affiliation(s)
- Betul Celik
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA;
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
| | - Andrés Felipe Leal
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Institute for the Study of Inborn Errors of Metabolism, Faculty of Science, Pontificia Universidad Javeriana, Bogotá 110231, Colombia
| | - Shunji Tomatsu
- Nemours Children’s Health, 1600 Rockland Rd., Wilmington, DE 19803, USA;
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, Philadelphia, PA 19144, USA
| |
Collapse
|
2
|
Akcaalan S, Akcan G, Tufan AC, Caglar C, Akcaalan Y, Akkaya M, Dogan M. Is tranexamic acid safe for the hip joint? NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1197-1207. [PMID: 37644283 DOI: 10.1007/s00210-023-02693-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 08/24/2023] [Indexed: 08/31/2023]
Abstract
To show the effects of tranexamic acid, which is a drug frequently used to control bleeding, on the hip joint and sciatic nerve in animal experiments. There were 15 rats in each of the 3 groups, with a total of 45 rats. Topical saline injections were applied to the first group, topical TXA injections to the second group, and intravenous (IV) TXA injections to the third group. In the samples taken from the hip joint 3 weeks later, femoral head cartilage, sciatic nerve, and joint capsule thicknesses were analyzed histologically. Statistically significantly more cartilage degradation was detected in the femoral head cartilage in both the IV and intraarticular TXA group when compared to the control group. The groups were also compared in terms of acetabular cartilage; however, no histological difference was found between the groups. It was seen that when the femoral head cartilage thickness (the average of the measurements made from 3 different points were used) was examined, the cartilage thickness in the topical TXA group was less when compared to the other 2 groups. However, this difference was determined to not be statistically significant. The data of the hip joint capsule thickness measurement, it was found that the capsule thickness in the topical TXA applied group was less when compared to the other 2 groups. However, this difference was not statistically significant. When the sciatic nerves in all 3 groups were compared, no different staining characteristics were found in the immunofluorescence examination. TXA, which is frequently used in orthopedic practice, shows negative effects on hip joint cartilage in both topical and intravenous application.
Collapse
Affiliation(s)
- Serhat Akcaalan
- Department of Orthopedics and Traumatology, Ankara Yıldırım Beyazıt University, 06800, Ankara, Turkey.
| | - Gulben Akcan
- Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, 06800, Ankara, Turkey
| | - Ahmet Cevik Tufan
- Department of Histology and Embryology, Ankara Yıldırım Beyazıt University, 06800, Ankara, Turkey
| | - Ceyhun Caglar
- Department of Orthopedics and Traumatology, Ankara City Hospital, 06800, Ankara, Turkey
| | | | - Mustafa Akkaya
- Department of Orthopedics and Traumatology, Ankara Yıldırım Beyazıt University, 06800, Ankara, Turkey
| | - Metin Dogan
- Department of Orthopedics and Traumatology, Ankara Yıldırım Beyazıt University, 06800, Ankara, Turkey
| |
Collapse
|
3
|
Wang Z, Zhu P, Liao B, You H, Cai Y. Effects and action mechanisms of individual cytokines contained in PRP on osteoarthritis. J Orthop Surg Res 2023; 18:713. [PMID: 37735688 PMCID: PMC10515001 DOI: 10.1186/s13018-023-04119-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 08/21/2023] [Indexed: 09/23/2023] Open
Abstract
Osteoarthritis (OA) is defined as a degenerative joint disease that can affect all tissues of the joint, including the articular cartilage, subchondral bone, ligaments capsule, and synovial membrane. The conventional nonoperative treatments are ineffective for cartilage repair and induce only symptomatic relief. Platelet-rich plasma (PRP) is a platelet concentrate derived from autologous whole blood with a high concentration of platelets, which can exert anti-inflammatory and regenerative effects by releasing multiple growth factors and cytokines. Recent studies have shown that PRP exhibits clinical benefits in patients with OA. However, high operational and equipment requirements greatly limit the application of PRP to OA treatment. Past studies have indicated that high-concentration PRP growth factors and cytokines may be applied as a commercial replacement for PRP. We reviewed the relevant articles to summarize the feasibility and mechanisms of PRP-based growth factors in OA. The available evidence suggests that transforming growth factor-α and β, platelet-derived growth factors, epidermal growth factor, insulin-like growth factor-1, and connective tissue growth factors might benefit OA, while vascular endothelial growth factor, tumor necrosis factor-α, angiopoietin-1, and stromal cell derived factor-1α might induce negative effects on OA. The effects of fibroblast growth factor, hepatocyte growth factor, platelet factor 4, and keratinocyte growth factor on OA remain uncertain. Thus, it can be concluded that not all cytokines released by PRP are beneficial, although the therapeutic action of PRP has a valuable potential to improve.
Collapse
Affiliation(s)
- Zhengchao Wang
- Department of Orthopedics, Wuhan Fourth Hospital, Wuhan, China
| | - Pengfei Zhu
- Department of Cardiovascular, Wuhan Fourth Hospital, Wuhan, China
- Department of Cardiovascular, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bokai Liao
- School of Chemistry and Chemical Engineering, Guangzhou University, Guangzhou, China
| | - Hongbo You
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University and Technology, Jiefang Avenue No.1095, Qiaokou District, Wuhan, 430030, Hubei Province, China.
| | - Yu Cai
- Department of Rehabilitation, Wuhan Fourth Hospital, Hanzheng Street No.473, Qiaokou District, Wuhan, 430000, Hubei Province, China.
| |
Collapse
|
4
|
Li J, Chen H, Cai L, Guo D, Zhang D, Zhou X, Xie J. SDF-1α Promotes Chondrocyte Autophagy through CXCR4/mTOR Signaling Axis. Int J Mol Sci 2023; 24:1710. [PMID: 36675225 PMCID: PMC9867011 DOI: 10.3390/ijms24021710] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 01/06/2023] [Accepted: 01/11/2023] [Indexed: 01/19/2023] Open
Abstract
SDF-1α, the most common isoform of stromal cell-derived factor 1, has shown vital effects in regulating chondrocyte proliferation, maturation, and chondrogenesis. Autophagy is a highly conserved biological process to help chondrocytes survive in harsh environments. However, the effect of SDF-1α on chondrocyte autophagy is still unknown. This study aims to investigate the effect of SDF-1α on chondrocyte autophagy and the underlying biomechanism. Transmission electron microscope assays and mRFP-GFP-LC3 adenovirus double label transfection assays were performed to detect the autophagic flux of chondrocytes. Western blots and immunofluorescence staining assays were used to detect the expression of autophagy-related proteins in chondrocytes. RNA sequencing and qPCR were conducted to assess changes in autophagy-related mRNA expression. SDF-1α upregulated the number of autophagosomes and autolysosomes in chondrocytes. It also increased the expression of autophagy-related proteins including ULK-1, Beclin-1 and LC3B, and decreased the expression of p62, an autophagy substrate protein. SDF-1α-mediated autophagy of chondrocytes required the participation of receptor CXCR4. Moreover, SDF-1α-enhanced autophagy of chondrocytes was through the inhibition of phosphorylation of mTOR signaling on the upstream of autophagy. Knockdown by siRNA and inhibition by signaling inhibitor further confirmed the importance of the CXCR4/mTOR signaling axis in SDF-1α-induced autophagy of chondrocytes. For the first time, this study elucidated that SDF-1α promotes chondrocyte autophagy through the CXCR4/mTOR signaling axis.
Collapse
Affiliation(s)
- Jiazhou Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Hao Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Lang Cai
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Daimo Guo
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Demao Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - Jing Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
- National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| |
Collapse
|
5
|
Selig M, Azizi S, Walz K, Lauer JC, Rolauffs B, Hart ML. Cell morphology as a biological fingerprint of chondrocyte phenotype in control and inflammatory conditions. Front Immunol 2023; 14:1102912. [PMID: 36860844 PMCID: PMC9968733 DOI: 10.3389/fimmu.2023.1102912] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Introduction Little is known how inflammatory processes quantitatively affect chondrocyte morphology and how single cell morphometric data could be used as a biological fingerprint of phenotype. Methods We investigated whether trainable high-throughput quantitative single cell morphology profiling combined with population-based gene expression analysis can be used to identify biological fingerprints that are discriminatory of control vs. inflammatory phenotypes. The shape of a large number of chondrocytes isolated from bovine healthy and human osteoarthritic (OA) cartilages was quantified under control and inflammatory (IL-1β) conditions using a trainable image analysis technique measuring a panel of cell shape descriptors (area, length, width, circularity, aspect ratio, roundness, solidity). The expression profiles of phenotypically relevant markers were quantified by ddPCR. Statistical analysis, multivariate data exploration, and projection-based modelling were used for identifying specific morphological fingerprints indicative of phenotype. Results Cell morphology was sensitive to both cell density and IL-1β. In both cell types, all shape descriptors correlated with expression of extracellular matrix (ECM)- and inflammatory-regulating genes. A hierarchical clustered image map revealed that individual samples sometimes responded differently in control or IL-1β conditions than the overall population. Despite these variances, discriminative projection-based modeling revealed distinct morphological fingerprints that discriminated between control and inflammatory chondrocyte phenotypes: the most essential morphological characteristics attributable to non-treated control cells was a higher cell aspect ratio in healthy bovine chondrocytes and roundness in OA human chondrocytes. In contrast, a higher circularity and width in healthy bovine chondrocytes and length and area in OA human chondrocytes indicated an inflammatory (IL-1β) phenotype. When comparing the two species/health conditions, bovine healthy and human OA chondrocytes exhibited comparable IL-1β-induced morphologies in roundness, a widely recognized marker of chondrocyte phenotype, and aspect ratio. Discussion Overall, cell morphology can be used as a biological fingerprint for describing chondrocyte phenotype. Quantitative single cell morphometry in conjunction with advanced methods for multivariate data analysis allows identifying morphological fingerprints that can discriminate between control and inflammatory chondrocyte phenotypes. This approach could be used to assess how culture conditions, inflammatory mediators, and therapeutic modulators regulate cell phenotype and function.
Collapse
Affiliation(s)
- Mischa Selig
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Saman Azizi
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Kathrin Walz
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Jasmin C Lauer
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany.,Faculty of Biology, University of Freiburg, Freiburg im Breisgau, Germany
| | - Bernd Rolauffs
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| | - Melanie L Hart
- G.E.R.N. Research Center for Tissue Replacement, Regeneration & Neogenesis, Department of Orthopedics and Trauma Surgery, Faculty of Medicine, Medical Center-Albert-Ludwigs-University of Freiburg, Freiburg im Breisgau, Germany
| |
Collapse
|
6
|
Xu Y, Duan L, Liu S, Yang Y, Qiao Z, Shi L. Long intergenic non-protein coding RNA 00707 regulates chondrocyte apoptosis and proliferation in osteoarthritis by serving as a sponge for microRNA-199-3p. Bioengineered 2022; 13:11137-11145. [PMID: 35485364 PMCID: PMC9208525 DOI: 10.1080/21655979.2022.2061287] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
It is known that long intergenic non-protein coding RNA 00707 (LINC00707) promotes lipopolysaccharide (LPS)-injury and microRNA-199-3p (miR-199-3p) regulates chondrocyte proliferation and apoptosis. Both processes participate in osteoarthritis (OA). We predicted that LINC00707 and miR-199-3p may interact with each other. Therefore, LINC00707 and miR-199-3p may interact with each other to participate in OA. In this study, the expression of LINC00707 and miR-199-3p in both OA and normal articular cartilage tissues was analyzed using RT-qPCR. The subcellular location of LINC00707 and its direct interaction with miR-199-3p were explored by nuclear fractionation assay, RNA pull-down assay and Luciferase reporter assay, respectively. The role of LINC00707 and miR-199-3p in regulating the expression of each other was analyzed using an overexpression assay, followed by RT-qPCR. The role of LINC00707 and miR-199-3p in regulating OA chondrocyte proliferation and apoptosis was analyzed by BrdU assay and cell apoptosis assay, respectively. OA tissues exhibited increased expression of LINC00707 and decreased expression of miR-199-3p. LINC00707 directly interacted with miR-199-3p in cytoplasm. However, LINC00707 and miR-199-3p overexpression failed to affect each other’s expression. LPS treatment increased LINC00707 expression and decreased miR-199-3p expression in OA chondrocyte. LINC00707 overexpression increased the apoptosis of OA chondrocytes induced by LPS and suppressed the proliferation of OA chondrocytes. Moreover, LINC00707 suppressed the role of miR-199-3p in enhancing cell proliferation and suppressing cell apoptosis. In conclusion, LINC00707 can be detected in cytoplasm and it may sponge miR-199-3p to regulate chondrocyte proliferation and apoptosis in OA.
Collapse
Affiliation(s)
- Yan Xu
- Department of Orthopedics, Xi'an Fifth Hospital, Xi'an, Shaanxi, PR China
| | - Liang Duan
- Department of Orthopedics, Shaanxi Provincial People's Hospital, Xian City, Shaanxi, PR China
| | - Shizhang Liu
- Department of Orthopedics, Shaanxi Provincial People's Hospital, Xian City, Shaanxi, PR China
| | - Yuanyuan Yang
- Editorial Board of Chinese Journal of Child Health Care, the Second Affiliated Hospital of Xi 'An Jiaotong University, Xian, Shaanxi, China
| | - Zhi Qiao
- Department of Orthopedics, Xi'an Fifth Hospital, Xi'an, Shaanxi, PR China
| | | |
Collapse
|
7
|
Farah H, Wijesinghe SN, Nicholson T, Alnajjar F, Certo M, Alghamdi A, Davis ET, Young SP, Mauro C, Jones SW. Differential Metabotypes in Synovial Fibroblasts and Synovial Fluid in Hip Osteoarthritis Patients Support Inflammatory Responses. Int J Mol Sci 2022; 23:ijms23063266. [PMID: 35328687 PMCID: PMC8950319 DOI: 10.3390/ijms23063266] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/02/2023] Open
Abstract
Changes in cellular metabolism have been implicated in mediating the activated fibroblast phenotype in a number of chronic inflammatory disorders, including pulmonary fibrosis, renal disease and rheumatoid arthritis. The aim of this study was therefore to characterise the metabolic profile of synovial joint fluid and synovial fibroblasts under both basal and inflammatory conditions in a cohort of obese and normal-weight hip OA patients. Furthermore, we sought to ascertain whether modulation of a metabolic pathway in OA synovial fibroblasts could alter their inflammatory activity. Synovium and synovial fluid was obtained from hip OA patients, who were either of normal-weight or obese and were undergoing elective joint replacement surgery. The synovial fluid metabolome was determined by 1H NMR spectroscopy. The metabolic profile of isolated synovial fibroblasts in vitro was characterised by lactate secretion, oxygen consumption rate (OCR) and extracellular acidification rate (ECAR) using the Seahorse XF Analyser. The effects of a small molecule pharmacological inhibitor and siRNA targeted at glutaminase-1 (GLS1) were assessed to probe the role of glutamine metabolism in OA synovial fibroblast function. Obese OA patient synovial fluid (n = 5) exhibited a different metabotype, compared to normal-weight patient fluid (n = 6), with significantly increased levels of 1, 3-dimethylurate, N-Nitrosodimethylamine, succinate, tyrosine, pyruvate, glucose, glycine and lactate, and enrichment of the glutamine-glutamate metabolic pathway, which correlated with increasing adiposity. In vitro, isolated obese OA fibroblasts exhibited greater basal lactate secretion and aerobic glycolysis, and increased mitochondrial respiration when stimulated with pro-inflammatory cytokine TNFα, compared to fibroblasts from normal-weight patients. Inhibition of GLS1 attenuated the TNFα-induced expression and secretion of IL-6 in OA synovial fibroblasts. These findings suggest that altered cellular metabolism underpins the inflammatory phenotype of OA fibroblasts, and that targeted inhibition of glutamine-glutamate metabolism may provide a route to reducing the pathological effects of joint inflammation in OA patients who are obese.
Collapse
Affiliation(s)
- Hussein Farah
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Susanne N. Wijesinghe
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Thomas Nicholson
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Fawzeyah Alnajjar
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Michelangelo Certo
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Abdullah Alghamdi
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Edward T. Davis
- The Royal Orthopaedic Hospital, NHS Foundation Trust, Bristol Road South, Northfield, Birmingham B31 2AP, UK;
| | - Stephen P. Young
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Claudio Mauro
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
| | - Simon W. Jones
- Institute of Inflammation and Ageing, MRC Versus Arthritis Centre for Musculoskeletal Ageing Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham B15 2TT, UK; (H.F.); (S.N.W.); (T.N.); (F.A.); (M.C.); (A.A.); (S.P.Y.); (C.M.)
- Correspondence:
| |
Collapse
|
8
|
Wen ZQ, Liu D, Zhang Y, Cai ZJ, Xiao WF, Li YS. G Protein-Coupled Receptors in Osteoarthritis: A Novel Perspective on Pathogenesis and Treatment. Front Cell Dev Biol 2021; 9:758220. [PMID: 34746150 PMCID: PMC8564363 DOI: 10.3389/fcell.2021.758220] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 09/27/2021] [Indexed: 11/30/2022] Open
Abstract
G protein-coupled receptors (GPCRs) are transmembrane receptor proteins that trigger numerous intracellular signaling pathways in response to the extracellular stimuli. The GPCRs superfamily contains enormous structural and functional diversity and mediates extensive biological processes. Until now, critical roles have been established in many diseases, including osteoarthritis (OA). Existing studies have shown that GPCRs play an important role in some OA-related pathogenesis, such as cartilage matrix degradation, synovitis, subchondral bone remodeling, and osteophyte formation. However, current pharmacological treatments are mostly symptomatic and there is a paucity of disease-modifying OA drugs so far. Targeting GPCRs is capable of inhibiting cartilage matrix degradation and synovitis and up-regulating cartilage matrix synthesis, providing a new therapeutic strategy for OA. In this review, we have comprehensively summarized the structures, biofunctions, and the novel roles of GPCRs in the pathogenesis and treatment of OA, which is expected to lay the foundation for the development of novel therapeutics against OA. Even though targeting GPCRs may ameliorate OA progression, many GPCRs-related therapeutic strategies are still in the pre-clinical stage and require further investigation.
Collapse
Affiliation(s)
- Ze-qin Wen
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Di Liu
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yi Zhang
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zi-jun Cai
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
| | - Wen-feng Xiao
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-sheng Li
- Department of Orthopedics, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
9
|
Oligonucleotide Therapies in the Treatment of Arthritis: A Narrative Review. Biomedicines 2021; 9:biomedicines9080902. [PMID: 34440106 PMCID: PMC8389545 DOI: 10.3390/biomedicines9080902] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 02/06/2023] Open
Abstract
Osteoarthritis (OA) and rheumatoid arthritis (RA) are two of the most common chronic inflammatory joint diseases, for which there remains a great clinical need to develop safer and more efficacious pharmacological treatments. The pathology of both OA and RA involves multiple tissues within the joint, including the synovial joint lining and the bone, as well as the articular cartilage in OA. In this review, we discuss the potential for the development of oligonucleotide therapies for these disorders by examining the evidence that oligonucleotides can modulate the key cellular pathways that drive the pathology of the inflammatory diseased joint pathology, as well as evidence in preclinical in vivo models that oligonucleotides can modify disease progression.
Collapse
|
10
|
Li J, Chen H, Zhang D, Xie J, Zhou X. The role of stromal cell-derived factor 1 on cartilage development and disease. Osteoarthritis Cartilage 2021; 29:313-322. [PMID: 33253889 DOI: 10.1016/j.joca.2020.10.010] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 02/05/2023]
Abstract
Stromal cell-derived factor 1 (SDF-1), also known as CXC motif chemokine ligand 12 (CXCL12), is recognized as a homeostatic cytokine with strong chemotactic potency. It plays an important role in physiological and pathological processes, such as the development of multiple tissues and organs, the regulation of cell distribution, and tumour metastasis. SDF-1 has two receptors, CXC chemokine receptor type 4 (CXCR4) and CXC chemokine receptor type 7 (CXCR7). SDF-1 affects the proliferation, survival, differentiation and maturation of chondrocytes by binding to CXCR4 on chondrocytes. Therefore, SDF-1 has been used as an exogenous regulatory target in many studies to explore the mechanism of cartilage development. SDF-1 is also a potential therapeutic target for osteoarthritis (OA) and rheumatoid arthritis (RA), because of its role in pathological initiation and regulation. In addition, SDF-1 shows potent capacity in the repair of cartilage defects by recruiting endogenous stem cells in a cartilage tissue engineering context. To summarize the specific role of SDF-1 on cartilage development and disease, all articles had been screened out in PubMed by May 30, 2020. The search was limited to studies published in English. Search terms included SDF-1; CXCL12; CXCR4; chondrocyte; cartilage; OA; RA, and forty-seven papers were studied. Besides, we reviewed references in the articles we searched to get additional relevant backgrounds. The review aims to conclude the current knowledge regarding the physiological and pathological role of SDF-1 on the cartilage and chondrocyte. More investigations are required to determine methods targeted SDF-1 to cartilage development and interventions to cartilage diseases.
Collapse
Affiliation(s)
- J Li
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - H Chen
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - D Zhang
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China
| | - J Xie
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - X Zhou
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Cariology and Endodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
11
|
Macfadyen MA, Daniel Z, Kelly S, Parr T, Brameld JM, Murton AJ, Jones SW. The commercial pig as a model of spontaneously-occurring osteoarthritis. BMC Musculoskelet Disord 2019; 20:70. [PMID: 30744620 PMCID: PMC6371556 DOI: 10.1186/s12891-019-2452-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2018] [Accepted: 02/01/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Preclinical osteoarthritis models where damage occurs spontaneously may better reflect the initiation and development of human osteoarthritis. The aim was to assess the commercial pig as a model of spontaneous osteoarthritis development by examining pain-associated behaviour, joint cartilage integrity, as well as the use of porcine cartilage explants and isolated chondrocytes and osteoblasts for ex vivo and in vitro studies. METHODS Female pigs (Large white x Landrace x Duroc) were examined at different ages from 6 weeks to 3-4 years old. Lameness was assessed as a marker of pain-associated behaviour. Femorotibial joint cartilage integrity was determined by chondropathy scoring and histological staining of proteoglycan. IL-6 production and proteoglycan degradation was assessed in cartilage explants and primary porcine chondrocytes by ELISA and DMMB assay. Primary porcine osteoblasts from damaged and non-damaged joints, as determined by chondropathy scoring, were assessed for mineralisation, proliferative and mitochondrial function as a marker of metabolic capacity. RESULTS Pigs aged 80 weeks and older exhibited lameness. Osteoarthritic lesions in femoral condyle and tibial plateau cartilage were apparent from 40 weeks and increased in severity with age up to 3-4 years old. Cartilage from damaged joints exhibited proteoglycan loss, which positively correlated with chondropathy score. Stimulation of porcine cartilage explants and primary chondrocytes with either IL-1β or visfatin induced IL-6 production and proteoglycan degradation. Primary porcine osteoblasts from damaged joints exhibited reduced proliferative, mineralisation, and metabolic capacity. CONCLUSION In conclusion, the commercial pig represents an alternative model of spontaneous osteoarthritis and an excellent source of tissue for in vitro and ex vivo studies.
Collapse
Affiliation(s)
- Mhairi A Macfadyen
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Zoe Daniel
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Sara Kelly
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Tim Parr
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - John M Brameld
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Biosciences, University of Nottingham, Sutton Bonington, UK
| | - Andrew J Murton
- MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Biosciences, University of Nottingham, Sutton Bonington, UK.,Metabolism Unit, Shriners Hospitals for Children, Galveston, TX, USA.,Department of Surgery, University of Texas Medical Branch, Galveston, TX, USA
| | - Simon W Jones
- Institute of Inflammation and Ageing, MRC-ARUK Centre for Musculoskeletal Ageing Research, School of Immunity, University of Birmingham, Birmingham, UK.
| |
Collapse
|
12
|
Baker J, Falconer AMD, Wilkinson DJ, Europe-Finner GN, Litherland GJ, Rowan AD. Protein kinase D3 modulates MMP1 and MMP13 expression in human chondrocytes. PLoS One 2018; 13:e0195864. [PMID: 29652915 PMCID: PMC5898748 DOI: 10.1371/journal.pone.0195864] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 03/31/2018] [Indexed: 01/02/2023] Open
Abstract
Many catabolic stimuli, including interleukin-1 (IL-1) in combination with oncostatin M (OSM), promote cartilage breakdown via the induction of collagen-degrading collagenases such as matrix metalloproteinase 1 (MMP1) and MMP13 in human articular chondrocytes. Indeed, joint diseases with an inflammatory component are characterised by excessive extracellular matrix (ECM) catabolism. Importantly, protein kinase C (PKC) signalling has a primary role in cytokine-induced MMP1/13 expression, and is known to regulate cellular functions associated with pathologies involving ECM remodelling. At present, substrates downstream of PKC remain undefined. Herein, we show that both IL-1- and OSM-induced phosphorylation of protein kinase D (PKD) in human chondrocytes is strongly associated with signalling via the atypical PKCι isoform. Consequently, inhibiting PKD activation with a pan-PKD inhibitor significantly reduced the expression of MMP1/13. Specific gene silencing of the PKD isoforms revealed that only PKD3 (PRKD3) depletion mirrored the observed MMP repression, indicative of the pharmacological inhibitor specifically affecting only this isoform. PRKD3 silencing was also shown to reduce serine phosphorylation of signal transducer and activator of transcription 3 (STAT3) as well as phosphorylation of all three mitogen-activated protein kinase groups. This altered signalling following PRKD3 silencing led to a significant reduction in the expression of the activator protein-1 (AP-1) genes FOS and JUN, critical for the induction of many MMPs including MMP1/13. Furthermore, the AP-1 factor activating transcription factor 3 (ATF3) was also reduced concomitant with the observed reduction in MMP13 expression. Taken together, we highlight an important role for PKD3 in the pro-inflammatory signalling that promotes cartilage destruction.
Collapse
Affiliation(s)
- Jonathan Baker
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Adrian M. D. Falconer
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - David J. Wilkinson
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - G. Nicholas Europe-Finner
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Gary J. Litherland
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Andrew D. Rowan
- Skeletal Research Group, Institute of Genetic Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
- * E-mail:
| |
Collapse
|
13
|
Nazari A, Khorramdelazad H, Hassanshahi G. Biological/pathological functions of the CXCL12/CXCR4/CXCR7 axes in the pathogenesis of bladder cancer. Int J Clin Oncol 2017; 22:991-1000. [PMID: 29022185 DOI: 10.1007/s10147-017-1187-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2017] [Accepted: 08/21/2017] [Indexed: 12/14/2022]
Abstract
CXC chemokine ligand 12 (CXCL12) is an important member of the CXC subfamily of chemokines, and has been extensively studied in various human body organs and systems, both in physiological and clinical states. Ligation of CXCL12 to CXCR4 and CXCR7 as its receptors on peripheral immune cells gives rise to pleiotropic activities. CXCL12 itself is a highly effective chemoattractant which conservatively attracts lymphocytes and monocytes, whereas there exists no evidence to show attraction for neutrophils. CXCL12 regulates inflammation, neo-vascularization, metastasis, and tumor growth, phenomena which are all pivotally involved in cancer development and further metastasis. Generation and secretion of CXCL12 by stromal cells facilitate attraction of cancer cells, acting through its cognate receptor, CXCR4, which is expressed by both hematopoietic and non-hematopoietic tumor cells. CXCR4 stimulates tumor progression by different mechanisms and is required for metastatic spread to organs where CXCL12 is expressed, thereby allowing tumor cells to access cellular niches, such as the marrow, which favor tumor cell survival and proliferation. It has also been demonstrated that CXCL12 binds to another seven-transmembrane G-protein receptor or G-protein-coupled receptor, namely CXCR7. These studies indicated critical roles for CXCR4 and CXCR7 mediation of tumor metastasis in several types of cancers, suggesting their contributions as biomarkers of tumor behavior as well as potential therapeutic targets. Furthermore, CXCL12 itself has the capability to stimulate survival and growth of neoplastic cells in a paracrine fashion. CXCL12 is a supportive chemokine for tumor neovascularization via attracting endothelial cells to the tumor microenvironment. It has been suggested that elevated protein and mRNA levels of CXCL12/CXCR4/CXCR7 are associated with human bladder cancer (BC). Taken together, mounting evidence suggests a role for CXCR4, CXCR7, and their ligand CXCL12 during the genesis of BC and its further development. However, a better understanding is still required before exploring CXCL12/CXCR4/CXCR7 targeting in the clinic.
Collapse
Affiliation(s)
- Alireza Nazari
- Department of Surgery, School of Medicine, Rafsanjan University of Medical Science, Rafsanjan, Iran.,Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Hossein Khorramdelazad
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Gholamhossein Hassanshahi
- Molecular Medicine Research Center, Rafsanjan University of Medical Sciences, Rafsanjan, Iran. .,Department of Immunology, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.
| |
Collapse
|
14
|
Lolli A, Penolazzi L, Narcisi R, van Osch GJVM, Piva R. Emerging potential of gene silencing approaches targeting anti-chondrogenic factors for cell-based cartilage repair. Cell Mol Life Sci 2017; 74:3451-3465. [PMID: 28434038 PMCID: PMC11107620 DOI: 10.1007/s00018-017-2531-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 04/14/2017] [Accepted: 04/19/2017] [Indexed: 12/18/2022]
Abstract
The field of cartilage repair has exponentially been growing over the past decade. Here, we discuss the possibility to achieve satisfactory regeneration of articular cartilage by means of human mesenchymal stem cells (hMSCs) depleted of anti-chondrogenic factors and implanted in the site of injury. Different types of molecules including transcription factors, transcriptional co-regulators, secreted proteins, and microRNAs have recently been identified as negative modulators of chondroprogenitor differentiation and chondrocyte function. We review the current knowledge about these molecules as potential targets for gene knockdown strategies using RNA interference (RNAi) tools that allow the specific suppression of gene function. The critical issues regarding the optimization of the gene silencing approach as well as the delivery strategies are discussed. We anticipate that further development of these techniques will lead to the generation of implantable hMSCs with enhanced potential to regenerate articular cartilage damaged by injury, disease, or aging.
Collapse
Affiliation(s)
- Andrea Lolli
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands.
| | - Letizia Penolazzi
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy
| | - Roberto Narcisi
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Gerjo J V M van Osch
- Department of Orthopaedics, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands
- Department of Otorhinolaryngology, Erasmus MC, University Medical Center, 3015 CN, Rotterdam, The Netherlands
| | - Roberta Piva
- Department of Biomedical and Specialty Surgical Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
15
|
Moazedi-Fuerst FC, Hofner M, Gruber G, Weinhaeusel A, Stradner MH, Angerer H, Peischler D, Lohberger B, Glehr M, Leithner A, Sonntagbauer M, Graninger WB. Epigenetic differences in human cartilage between mild and severe OA. J Orthop Res 2014; 32:1636-45. [PMID: 25212754 DOI: 10.1002/jor.22722] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 07/24/2014] [Indexed: 02/04/2023]
Abstract
The development of osteoarthritis (OA) depends on genetic and environmental factors, which influence the biology of the chondrocyte via epigenetic regulation. Changes within the epigenome might lead the way to discovery of new pathogenetic pathways. We performed a genome-wide methylation screening to identify potential differences between paired mild and severe osteoarthritic human cartilage. Sixteen female patients suffering from OA underwent total knee joint replacement. Cartilage specimens collected from corresponding macroscopically undamaged and from damaged areas were processed for DNA extraction and histology to evaluate the histological grading of the disease. Paired specimens were analysed for the methylation status of the whole genome using human promoter microarrays (Agilent, Santa Clara, CA). Selected target genes were then validated via methylation-specific qPCR. One thousand two hundred and fourteen genetic targets were identified differentially methylated between mild and severe OA. One thousand and seventy of these targets were found hypermethylated and 144 hypomethylated. The descriptive analysis of these genes by Gene Ontology (GO), KEGG pathway and protein domain analyses points to pathways of development and differentiation. We identified a list of genes which are differently methylated in mild and severe OA cartilage. Within the pathways of growth and development new therapeutic targets might arise by improving our understanding of pathogenetic mechanisms in OA.
Collapse
|
16
|
Abstract
Herein, we review the regulation of differentiation of the growth plate chondrocytes by G-proteins. In connection with this, we summarize the current knowledge regarding each family of G-protein α subunit, specifically, Gα(s), Gα(q/11), Gα(12/13), and Gα(i/o). We discuss different mechanisms involved in chondrocyte differentiation downstream of G-proteins and different G-protein-coupled receptors (GPCRs) activating G-proteins in the epiphyseal chondrocytes. We conclude that among all G-proteins and GPCRs expressed by chondrocytes, Gα(s) has the most important role and prevents premature chondrocyte differentiation. Receptor for parathyroid hormone (PTHR1) appears to be the major activator of Gα(s) in chondrocytes and ablation of either one leads to accelerated chondrocyte differentiation, premature fusion of the postnatal growth plate, and ultimately short stature.
Collapse
Affiliation(s)
- Andrei S Chagin
- Department of Physiology and PharmacologyKarolinska Institutet, Nanna Svartz Vagen 2, Stockholm 17177, SwedenEndocrine UnitMassachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114-2696, USA
| | - Henry M Kronenberg
- Department of Physiology and PharmacologyKarolinska Institutet, Nanna Svartz Vagen 2, Stockholm 17177, SwedenEndocrine UnitMassachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114-2696, USA
| |
Collapse
|
17
|
Ho IAW, Yulyana Y, Sia KC, Newman JP, Guo CM, Hui KM, Lam PYP. Matrix metalloproteinase-1-mediated mesenchymal stem cell tumor tropism is dependent on crosstalk with stromal derived growth factor 1/C-X-C chemokine receptor 4 axis. FASEB J 2014; 28:4359-68. [PMID: 25271298 DOI: 10.1096/fj.14-252551] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Human bone marrow-derived mesenchymal stem cells (MSCs) have the unique ability to home toward injuries or tumor sites. We have previously shown that the tumor-tropic property is dependent on the intrinsic expression and activity of the matrix remodeling gene, matrix metalloproteinase 1 (MMP-1). Herein, crosstalk between MMP-1/protease activated receptor 1 (PAR-1) and the G-protein coupled receptor stromal-derived growth factor 1 (SDF-1)/C-X-C chemokine receptor 4 (CXCR-4) in facilitating cell migration was investigated. Gain-of-function and RNA interference (RNAi) technology were used to evaluate the interplay between the key players. The downstream effect on the tumor-tropic migration of MSCs was investigated using modified Boyden chamber assay. Neutralizing PAR-1 activation using monoclonal antibody and targeted knockdown of MMP-1 using RNAi resulted in decreased expression of SDF-1, which was not observed in control-RNAi-transfected cells. Overexpression of CXCR-4 failed to promote MSC migration; the percentage of migrated cells toward tumor cell conditioned medium was similar to the vector-transduced and the CXCR-4-transduced MSCs. Furthermore, inhibition of SDF-1/CXCR-4 signaling using AMD3100 reduced MSC migration through the deregulation of MMP-1 promoter activities, protein expression, and metalloproteinase activity. Collectively, our results showed that MMP-1-mediated MSC tumor tropism is dependent on crosstalk with the SDF-1/CXCR-4 axis.
Collapse
Affiliation(s)
| | | | | | | | - Chang M Guo
- Department of Orthopedics, Singapore General Hospital, Singapore
| | - Kam M Hui
- Bek Chai Heah Laboratory of Cancer Genomics, Cellular and Molecular Research Division, Humphrey Oei Institute of Cancer Research, National Cancer Centre, Singapore; Cancer and Stem Cells Biology Program, Duke-National University of Singapore Graduate Medical School, and Department of Biochemistry and Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), Singapore
| | - Paula Y P Lam
- Laboratory of Cancer Gene Therapy and Cancer and Stem Cells Biology Program, Duke-National University of Singapore Graduate Medical School, and Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; and
| |
Collapse
|
18
|
Villalvilla A, Gomez R, Roman-Blas JA, Largo R, Herrero-Beaumont G. SDF-1 signaling: a promising target in rheumatic diseases. Expert Opin Ther Targets 2014; 18:1077-87. [DOI: 10.1517/14728222.2014.930440] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
19
|
Bateman JF, Rowley L, Belluoccio D, Chan B, Bell K, Fosang AJ, Little CB. Transcriptomics of wild-type mice and mice lacking ADAMTS-5 activity identifies genes involved in osteoarthritis initiation and cartilage destruction. ACTA ACUST UNITED AC 2013; 65:1547-60. [PMID: 23436205 DOI: 10.1002/art.37900] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2012] [Accepted: 02/05/2013] [Indexed: 01/10/2023]
Abstract
OBJECTIVE To identify changes in gene expression in mice with osteoarthritis (OA) in order to explore the mechanisms of the disease. METHODS Gene expression profiling was performed in cartilage from mice with surgically induced OA. We used wild-type (WT) mice and Adamts5Δcat mice, in which ADAMTS-5 activity is lacking and aggrecan loss and cartilage erosion are inhibited, to distinguish gene expression changes that are independent of ADAMTS-5 activity and cartilage breakdown. Mechanical instability was introduced into the knee joints of 10-week-old male mice via surgical destabilization of the medial meniscus (DMM). Cartilage from the developing lesion in the destabilized medial meniscus and corresponding regions in sham-operated joints was harvested by microdissection at 1, 2, and 6 weeks postsurgery, and RNA was extracted, amplified, and hybridized to whole-genome microarrays. RESULTS Several previously identified OA-related genes, including Ptgs2, Crlf1, and Inhba, and novel genes, such as Phdla2 and Il11, were up-regulated in both WT mice and Adamts5Δcat mice, indicating that they are independent of ADAMTS-5 activity. The altered expression of other genes, including Col10a1, the sentinel marker of cartilage hypertrophy, and Wnt/β-catenin pathway genes, required ADAMTS-5 activity. Cell death pathway genes were dysregulated, and Tp53, Foxo4, and Xbp1 endoplasmic reticulum-stress transcriptional networks were activated. Analysis of degradome genes identified up-regulation of many proteases, including Mmp3, Capn2, and the novel cartilage proteases Prss46 and Klk8. Comparison with other studies identified 16 genes also dysregulated in rat and human OA as priorities for study. CONCLUSION We have identified, for the first time, several genes that have an ADAMTS-5-independent role in OA, identifying them as possible OA initiation candidates. This work provides new insights into the sequence of gene dysregulation and the molecular basis of cartilage destruction in OA.
Collapse
Affiliation(s)
- John F Bateman
- Murdoch Childrens Research Institute and University of Melbourne, Parkville, Victoria, Australia.
| | | | | | | | | | | | | |
Collapse
|
20
|
Xue TC, Chen RX, Ren ZG, Zou JH, Tang ZY, Ye SL. Transmembrane receptor CXCR7 increases the risk of extrahepatic metastasis of relatively well-differentiated hepatocellular carcinoma through upregulation of osteopontin. Oncol Rep 2013; 30:105-10. [PMID: 23636305 DOI: 10.3892/or.2013.2442] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 02/14/2013] [Indexed: 12/29/2022] Open
Abstract
Recurrence and metastasis are the main obstacles to improving the survival of patients with post-resective hepato-cellular carcinoma (HCC). Our previous study suggests a critical role of CXCR7 in the metastasis of HCC. In the present study, the effect of CXCR7 as a risk factor for metastasis of HCC was evaluated. Immunohistochemical assay was performed on tissue microarrays based on HCC with extrahepatic metastases after hepatectomy. Two categories based on staining scores were used to evaluate the risk effect of CXCR7, respectively. The effect of CXCR7 on osteopontin (OPN) was explored by RNA interference. Based on the results, in both categories, highly expressed CXCR7 was a dependent risk factor for extrahepatic metastasis because of the potential association with relatively good cell differentiation. Stratification analyses indicated that CXCR7 was a strong independent risk factor (OR, 3.40; 95% CI, 1.07-18.84; P=0.038 in category 1 and OR, 6.40; 95% CI, 1.64-24.92; P=0.007 in category 2, respectively) in patients with Edmondson grade 1/2. Furthermore, CXCR7 correlated well and positively with expression of OPN (P=0.019 and P<0.001 in two categories, respectively) in HCC cases with Edmondson grade 1/2. Immunocytochemistry and RT-PCR demonstrated downregulation of OPN in a highly metastatic HCC cell line following knockdown of CXCR7. Taken together, these findings suggest that high expression of CXCR7 increases the risk of metastasis in post-resective HCC patients with relatively good differentiated tumors, potentially through upregulation of OPN. This group of patients may acquire a survival benefit from early detection and treatment of recurrence and metastasis.
Collapse
Affiliation(s)
- Tong-Chun Xue
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | | | | | | | | | | |
Collapse
|
21
|
Akimoto K, Kimura K, Nagano M, Takano S, To'a Salazar G, Yamashita T, Ohneda O. Umbilical cord blood-derived mesenchymal stem cells inhibit, but adipose tissue-derived mesenchymal stem cells promote, glioblastoma multiforme proliferation. Stem Cells Dev 2013; 22:1370-86. [PMID: 23231075 DOI: 10.1089/scd.2012.0486] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stem cells (MSCs) possess self-renewal and multipotential differentiation abilities, and they are thought to be one of the most reliable stem cell sources for a variety of cell therapies. Recently, cell therapy using MSCs has been studied as a novel therapeutic approach for cancers that show refractory progress and poor prognosis. MSCs from different tissues have different properties. However, the effect of different MSC properties on their application in anticancer therapies has not been thoroughly investigated. In this study, to characterize the anticancer therapeutic application of MSCs from different sources, we established two different kinds of human MSCs: umbilical cord blood-derived MSCs (UCB-MSCs) and adipose-tissue-derived MSCs (AT-MSCs). We used these MSCs in a coculture assay with primary glioblastoma multiforme (GBM) cells to analyze how MSCs from different sources can inhibit GBM growth. We found that UCB-MSCs inhibited GBM growth and caused apoptosis, but AT-MSCs promoted GBM growth. Terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick-end labeling assay clearly demonstrated that UCB-MSCs promoted apoptosis of GBM via tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). TRAIL was expressed more highly by UCB-MSCs than by AT-MSCs. Higher mRNA expression levels of angiogenic factors (vascular endothelial growth factor, angiopoietin 1, platelet-derived growth factor, and insulin-like growth factor) and stromal-derived factor-1 (SDF-1/CXCL12) were observed in AT-MSCs, and highly vascularized tumors were developed when AT-MSCs and GBM were cotransplanted. Importantly, CXCL12 inhibited TRAIL activation of the apoptotic pathway in GBM, suggesting that AT-MSCs may support GBM development in vivo by at least two distinct mechanisms-promoting angiogenesis and inhibiting apoptosis. The opposite effects of AT-MSCs and UCB-MSCs on GBM clearly demonstrate that differences must be considered when choosing a stem cell source for safety in clinical application.
Collapse
Affiliation(s)
- Keiko Akimoto
- Department of Regenerative Medicine, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Japan
| | | | | | | | | | | | | |
Collapse
|
22
|
Sánchez-Martín L, Sánchez-Mateos P, Cabañas C. CXCR7 impact on CXCL12 biology and disease. Trends Mol Med 2012; 19:12-22. [PMID: 23153575 DOI: 10.1016/j.molmed.2012.10.004] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Revised: 09/20/2012] [Accepted: 10/11/2012] [Indexed: 12/14/2022]
Abstract
It is known that the chemokine receptor CXCR7 (RDC1) can be engaged by both chemokines CXCL12 (SDF-1) and CXCL11 (I-TAC), but the exact expression pattern and function of CXCR7 is controversial. CXCR7 expression seems to be enhanced during pathological inflammation and tumor development, and emerging data suggest this receptor is an attractive therapeutic target for autoimmune diseases and cancer. CXCR7/CXCR4 heterodimerization, β-arrestin-mediated signaling, and modulation of CXCL12 responsiveness by CXCR7 suggest that the monogamous CXCR4/CXCL12 signaling axis is an oversimplified model that needs to be revisited. Consequently, research into CXCR7 biology is of great interest and further studies are warranted. This review summarizes recent findings about the CXCR7 receptor and analyses its impact on understanding the roles of CXCL12 biology in health and disease.
Collapse
Affiliation(s)
- Lorena Sánchez-Martín
- Departamento de Biología Celular e Inmunología, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid 28049, Spain.
| | | | | |
Collapse
|
23
|
Scholten DJ, Canals M, Maussang D, Roumen L, Smit MJ, Wijtmans M, de Graaf C, Vischer HF, Leurs R. Pharmacological modulation of chemokine receptor function. Br J Pharmacol 2012; 165:1617-1643. [PMID: 21699506 DOI: 10.1111/j.1476-5381.2011.01551.x] [Citation(s) in RCA: 193] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
G protein-coupled chemokine receptors and their peptidergic ligands are interesting therapeutic targets due to their involvement in various immune-related diseases, including rheumatoid arthritis, multiple sclerosis, inflammatory bowel disease, chronic obstructive pulmonary disease, HIV-1 infection and cancer. To tackle these diseases, a lot of effort has been focused on discovery and development of small-molecule chemokine receptor antagonists. This has been rewarded by the market approval of two novel chemokine receptor inhibitors, AMD3100 (CXCR4) and Maraviroc (CCR5) for stem cell mobilization and treatment of HIV-1 infection respectively. The recent GPCR crystal structures together with mutagenesis and pharmacological studies have aided in understanding how small-molecule ligands interact with chemokine receptors. Many of these ligands display behaviour deviating from simple competition and do not interact with the chemokine binding site, providing evidence for an allosteric mode of action. This review aims to give an overview of the evidence supporting modulation of this intriguing receptor family by a range of ligands, including small molecules, peptides and antibodies. Moreover, the computer-assisted modelling of chemokine receptor-ligand interactions is discussed in view of GPCR crystal structures. Finally, the implications of concepts such as functional selectivity and chemokine receptor dimerization are considered.
Collapse
Affiliation(s)
- D J Scholten
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - M Canals
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - D Maussang
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - L Roumen
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - M J Smit
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - M Wijtmans
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - C de Graaf
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - H F Vischer
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| | - R Leurs
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Science, VU University Amsterdam, Amsterdam, the Netherlands
| |
Collapse
|
24
|
Structural modelling and dynamics of proteins for insights into drug interactions. Adv Drug Deliv Rev 2012; 64:323-43. [PMID: 22155026 DOI: 10.1016/j.addr.2011.11.011] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2011] [Revised: 11/17/2011] [Accepted: 11/24/2011] [Indexed: 12/27/2022]
Abstract
Proteins are the workhorses of biomolecules and their function is affected by their structure and their structural rearrangements during ligand entry, ligand binding and protein-protein interactions. Hence, the knowledge of protein structure and, importantly, the dynamic behaviour of the structure are critical for understanding how the protein performs its function. The predictions of the structure and the dynamic behaviour can be performed by combinations of structure modelling and molecular dynamics simulations. The simulations also need to be sensitive to the constraints of the environment in which the protein resides. Standard computational methods now exist in this field to support the experimental effort of solving protein structures. This review presents a comprehensive overview of the basis of the calculations and the well-established computational methods used to generate and understand protein structure and function and the study of their dynamic behaviour with the reference to lung-related targets.
Collapse
|
25
|
Wijtmans M, Maussang D, Sirci F, Scholten DJ, Canals M, Mujić-Delić A, Chong M, Chatalic KLS, Custers H, Janssen E, de Graaf C, Smit MJ, de Esch IJP, Leurs R. Synthesis, modeling and functional activity of substituted styrene-amides as small-molecule CXCR7 agonists. Eur J Med Chem 2012; 51:184-92. [PMID: 22424612 DOI: 10.1016/j.ejmech.2012.02.041] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2011] [Revised: 02/18/2012] [Accepted: 02/20/2012] [Indexed: 12/22/2022]
Abstract
The chemokine receptor CXCR7 is an atypical G protein-coupled receptor as it preferentially signals through the β-arrestin pathway rather than through G proteins. CXCR7 is thought to be of importance in cancer and the development of CXCR7-targeting ligands is of huge importance to further elucidate the pharmacology and the therapeutic potential of CXCR7. In the present study, we synthesized 24 derivatives based on a compound scaffold patented by Chemocentryx and obtained CXCR7 ligands with pK(i) values ranging from 5.3 to 8.1. SAR studies were supported by computational 3D Fingerprint studies, revealing several important affinity descriptors. Two key compounds (29 and 30, VUF11207 and VUF11403) were found to be high-potency ligands that induce recruitment of β-arrestin2 and subsequent internalization of CXCR7, making them important tool compounds in future CXCR7 research.
Collapse
Affiliation(s)
- Maikel Wijtmans
- Leiden/Amsterdam Center for Drug Research, Division of Medicinal Chemistry, Faculty of Sciences, VU University Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
CXCR7 mediated Giα independent activation of ERK and Akt promotes cell survival and chemotaxis in T cells. Cell Immunol 2011; 272:230-41. [PMID: 22070874 DOI: 10.1016/j.cellimm.2011.09.015] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2011] [Revised: 09/11/2011] [Accepted: 09/13/2011] [Indexed: 11/21/2022]
Abstract
Chemokine receptors CXCR7 and CXCR4 bind to the same ligand stromal cell derived factor-1alpha (SDF-1α/CXCL12). We assessed the downstream signaling pathways mediated by CXCL12-CXCR7 interaction in Jurkat T cells. All experiments were carried out after functionally blocking the CXCR4 receptor. CXCL12, on binding CXCR7, induced phosphorylation of extra cellular regulated protein kinases (ERK 1/2) and Akt. Selective inhibition of each signal demonstrated that phosphorylated ERK 1/2 is essential for chemotaxis and survival of T cells whereas activation of Akt promotes only cell survival. Another interesting finding of this study is that CXCL12-CXCR7 interaction under normal physiological conditions does not activate the p38 pathway. Furthermore, we observed that the CXCL12 signaling via CXCR7 is Giα independent. Our findings suggest that CXCR7 promotes cell survival and does not induce cell death in T cells. The CXCL12 signaling via CXCR7 may be crucial in determining the fate of the activated T cells.
Collapse
|
27
|
Xue TC, Chen RX, Han D, Chen J, Xue Q, Gao DM, Sun RX, Tang ZY, Ye SL. Down-regulation of CXCR7 inhibits the growth and lung metastasis of human hepatocellular carcinoma cells with highly metastatic potential. Exp Ther Med 2011; 3:117-123. [PMID: 22969855 DOI: 10.3892/etm.2011.358] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Accepted: 08/08/2011] [Indexed: 12/13/2022] Open
Abstract
CXCR7, a recently identified chemokine receptor, has been implicated in directing cancer metastasis. In the present study, the potential roles of CXCR7 in the growth and metastasis of hepatocellular carcinoma (HCC) were evaluated. A chemokine receptor gene chip was used to compare the expression of CXCR7 in HCC cell lines with different metastatic potential. Effects of targeting CXCR7 by RNA interference (RNAi) on the proliferation and metastasis of HCCLM3 cells were observed in vitro and in vivo. CXCR7 expression in 116 specimens from patients with or without metastatic HCC was assessed by tissue microarray. As a result, the gene chip showed that expression of CXCR7 was significantly higher in the highly metastatic HCCLM3 cells, which was confirmed by real-time RT-PCR and Western blotting. Chemotaxis assays showed that HCCLM3 cells responded to SDF-1α from 1 to 100 μg/l and lung extractions (1 g/l). Furthermore, down-regulation of CXCR7 in HCCLM3 cells by RNAi inhibited the proliferation and invasion of tumor cells in vitro. Moreover, CXCR7 knockdown significantly reduced the activity of matrix metalloproteinase-2 and matrix metalloproteinase-9. RNAi of CXCR7 in the HCCLM3 cells also decreased the growth of tumors and the number of lung metastases in nude mice. The tissue microarray showed that HCCs with high expression of CXCR7 were prone to metastasize to the lung. These findings suggest that CXCR7 plays critical roles in the growth and metastasis of HCC. RNAi of CXCR7 inhibits the growth and invasion of tumor cells, which indicates that CXCR7 may be a potential molecular target for use in HCC therapy.
Collapse
Affiliation(s)
- Tong-Chun Xue
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Sun X, Cheng G, Hao M, Zheng J, Zhou X, Zhang J, Taichman RS, Pienta KJ, Wang J. CXCL12 / CXCR4 / CXCR7 chemokine axis and cancer progression. Cancer Metastasis Rev 2011; 29:709-22. [PMID: 20839032 DOI: 10.1007/s10555-010-9256-x] [Citation(s) in RCA: 561] [Impact Index Per Article: 40.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Chemokines, small pro-inflammatory chemoattractant cytokines that bind to specific G-protein-coupled seven-span transmembrane receptors, are major regulators of cell trafficking and adhesion. The chemokine CXCL12 (also called stromal-derived factor-1) is an important α-chemokine that binds primarily to its cognate receptor CXCR4 and thus regulates the trafficking of normal and malignant cells. For many years, it was believed that CXCR4 was the only receptor for CXCL12. Yet, recent work has demonstrated that CXCL12 also binds to another seven-transmembrane span receptor called CXCR7. Our group and others have established critical roles for CXCR4 and CXCR7 on mediating tumor metastasis in several types of cancers, in addition to their contributions as biomarkers of tumor behavior as well as potential therapeutic targets. Here, we review the current concepts regarding the role of CXCL12 / CXCR4 / CXCR7 axis activation, which regulates the pattern of tumor growth and metastatic spread to organs expressing high levels of CXCL12 to develop secondary tumors. We also summarize recent therapeutic approaches to target these receptors and/or their ligands.
Collapse
Affiliation(s)
- Xueqing Sun
- Department of Biochemistry and Molecular & Cell Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Medical Science, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
The role of stromal-derived factor-1--CXCR7 axis in development and cancer. Eur J Pharmacol 2009; 625:31-40. [PMID: 19835865 DOI: 10.1016/j.ejphar.2009.04.071] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 03/24/2009] [Accepted: 04/29/2009] [Indexed: 01/07/2023]
Abstract
Cancer metastasis is a major clinical problem that contributes to unsuccessful therapy. Augmenting evidence indicates that metastasizing cancer cells employ several mechanisms that are involved in developmental trafficking of normal stem cells. Stromal-derived factor-1 (SDF-1) is an important alpha-chemokine that binds to the G-protein-coupled seven-transmembrane span CXCR4. The SDF-1-CXCR4 axis regulates trafficking of normal and malignant cells. SDF-1 is an important chemoattractant for a variety of cells including hematopoietic stem/progenitor cells. For many years, it was believed that CXCR4 was the only receptor for SDF-1. However, several reports recently provided evidence that SDF-1 also binds to another seven-transmembrane span receptor called CXCR7, sharing this receptor with another chemokine family member called Interferon-inducible T-cell chemoattractant (I-TAC). Thus, with CXCR7 identified as a new receptor for SDF-1, the role of the SDF-1-CXCR4 axis in regulating several biological processes becomes more complex. Based on the available literature, this review addresses the biological significance of SDF-1's interaction with CXCR7, which may act as a kind of decoy or signaling receptor depending on cell type. Augmenting evidence suggests that CXCR7 is involved in several aspects of tumorogenesis and could become an important target for new anti-metastatic and anti-cancer drugs.
Collapse
|
30
|
Cubedo N, Cerdan E, Sapede D, Rossel M. CXCR4 and CXCR7 cooperate during tangential migration of facial motoneurons. Mol Cell Neurosci 2009; 40:474-84. [PMID: 19340934 DOI: 10.1016/j.mcn.2009.01.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Migration of facial motoneurons in the zebrafish hindbrain depends on SDF1/CXCL12 signaling. Recent studies demonstrated that SDF1 can bind two chemokine receptors, CXCR4 and CXCR7. Here we explore the expression and function of the cxcr7b gene in zebrafish hindbrain development. By the time cxcr4b-expressing motoneurons migrate from rhombomere (r) r4 to r6, expression of cxcr7b is rapidly restricted to the ventral part of r5. Inactivation of either cxcr7b or cxcr4b impairs motoneuron migration, with however different phenotypes. Facial motoneurons preferentially accumulate in r5 in cxcr7b morphant embryos, while they are distributed between r4, r5 and r6 in cxcr4b morphants. Simultaneous inactivation of both receptors leads to yet a third phenotype, with motoneurons mostly distributed between r4 and r5. The latter phenotype resembles that of sdf1a morphant embryos. Double inactivation of sdf1a and cxcr7b indeed did not lead to a complete arrest of migration but rather to a partial rescue of r5 arrest of motoneuron migration. This result is in accordance with the functional hypothesis that SDF1 might interact with CXCR7 and that they have an antagonistic effect within r5. The ectopic expression of a truncated CXCR7 receptor leads to a motoneuron migration defect. Altogether, we show that CXCR7 is required, for proper tangential migration of facial motoneurons, by determining a permissive migration pathway through r5.
Collapse
Affiliation(s)
- Nicolas Cubedo
- Institut National de la Santé et de la Recherche Médicale U881-cc103, Pl E Bataillon 34095, Montpellier-France.
| | | | | | | |
Collapse
|
31
|
Yoshida D, Nomura R, Teramoto A. Signalling pathway mediated by CXCR7, an alternative chemokine receptor for stromal-cell derived factor-1α, in AtT20 mouse adrenocorticotrophic hormone-secreting pituitary adenoma cells. J Neuroendocrinol 2009; 21:481-8. [PMID: 19302186 DOI: 10.1111/j.1365-2826.2009.01867.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Stromal cell-derived factor (SDF)-1 and its receptor, CXCR4, have been identified in both neurones and glia of many brain areas. Previous studies have mainly focused on the role of SDF-1 and CXCR4 in modulating the hypothalamic-pituitary axis and their possible involvement in the development of pituitary adenomas. An alternative SDF-1 receptor, CXCR7, has recently been identified, but it has not been studied in the context of pituitary adenomas. The present study aimed to investigate the distribution and function of CXCR7 in pituitary adenomas. The expression of CXCR7, normalised to β-actin, was assessed by tissue microarray analysis of 62 adenomas, including 23 growth hormone (GH)-producing adenomas, 22 nonfunctioning adenomas, seven prolactin (PRL)-producing adenomas, six adrenocorticotrophic hormone-producing adenomas and four thyroid-stimulating hormone-producing adenomas. In vitro functional studies used RNA interference (RNAi) and cDNA microarray analysis to evaluate the CXCR7 signalling pathway in AtT-20 mouse pituitary adenoma cells treated with recombinant mouse SDF-1α and transfected with RNAi against Cxcr7 or control RNAi. In tissue microarray analysis, prominent expression of CXCR7 was observed in GH-producing adenomas and PRL-producing adenomas, and in macroadenomas (P < 0.05). Intracellular signalling via CXCR7 up-regulated Bub1, Cdc29 and Ccnb1, and down-regulated Asns, Gpt, Pycr1, Cars and Dars. The present study demonstrates that the SDF-1α ⁄ CXCR7 signalling pathway regulates genes involved in cell cycle control, amino acid metabolism and ligase activity, which comprise targets that are distinct from those of CXCR4.
Collapse
Affiliation(s)
- D Yoshida
- Department of Neurosurgery, Nippon Medical School, Tokyo, Japan.
| | | | | |
Collapse
|
32
|
Differential expression of RDC1/CXCR7 in the human placenta. J Clin Immunol 2008; 29:379-86. [PMID: 18956235 DOI: 10.1007/s10875-008-9258-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Accepted: 10/07/2008] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Chemokine receptor expression by human trophoblast and other placental cells have important implications for understanding the regulation of placental growth, development, and their role in maternofetal HIV transmission. CXCR7, now a deorphanized G protein coupled receptor that has been recently shown to bind to the ligands ITAC and CXCL12 has been proposed to act as a co-receptor for HIV-1, HIV-2, and SIV strains. The differential expression of CXCR7 in the human placenta is not yet reported. METHODS The expression of CXCR7 was studied in 45 different human placental tissues, of which 20 were from early placental tissues (8-10 week old) obtained from medically terminated pregnancies and 25 were placenta from normal term deliveries. RESULTS Immunohistochemistry and RT-PCR analysis revealed a greater expression of CXCR7 in term human placenta as compared to the early stage. This was further confirmed by real-time PCR. CONCLUSION Our study reveals, for the first time, the differential expression of CXCR7 in early (8-10 weeks) and term human placenta. The precise role of CXCR7 in the human placenta needs to be determined. HIV vertical transmission is reported to occur mainly during the end stages of pregnancy. Our finding of increased CXCR7 expression in the term human placenta therefore warrants future studies to assess its role in the vertical transmission of HIV-1.
Collapse
|
33
|
Gerrits H, van Ingen Schenau DS, Bakker NEC, van Disseldorp AJM, Strik A, Hermens LS, Koenen TB, Krajnc-Franken MAM, Gossen JA. Early postnatal lethality and cardiovascular defects in CXCR7-deficient mice. Genesis 2008; 46:235-45. [PMID: 18442043 DOI: 10.1002/dvg.20387] [Citation(s) in RCA: 114] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
CXCR7 is a G-protein coupled receptor that was recently deorphanized and shown to have SDF1 and I-TAC as high affinity ligands. Here we describe the characterization of CXCR7-deficient mice that were generated to further investigate the function of this receptor in vivo. Expression analysis using a LacZ reporter knockin revealed that postnatally Cxcr7 was specifically expressed in cardiomyocytes, vascular endothelial cells of the lung and heart, the cerebral cortex and in osteocytes of the bone. Adult tissues revealed high expression in cardiomyocytes and osteocytes. The observation that 70% of the Cxcr7-/- mice died in the first week after birth coincides with expression of Cxcr7 in vascular endothelial cells and in cardiomyocytes. An important role of CXCR7 in the cardiovascular system was further supported by the observation that hearts of the Cxcr7-/- mice were enlarged, showed myocardial degeneration and fibrosis of postnatal origin, and hyperplasia of embryonic origin. Despite high expression in osteocytes no apparent bone phenotype was observed, neither in combination with ovariectomy nor orchidectomy. Thus as CXCR7 does not seem to play an important role in bone our data indicate an important function of CXCR7 in the cardiovascular system during multiple steps of development.
Collapse
Affiliation(s)
- Han Gerrits
- N.V. Organon, part of Schering-Plough Corporation, Target Discovery, Molenstraat 110, 5340 BH Oss, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Andreas K, Lübke C, Häupl T, Dehne T, Morawietz L, Ringe J, Kaps C, Sittinger M. Key regulatory molecules of cartilage destruction in rheumatoid arthritis: an in vitro study. Arthritis Res Ther 2008; 10:R9. [PMID: 18205922 PMCID: PMC2374452 DOI: 10.1186/ar2358] [Citation(s) in RCA: 87] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2007] [Revised: 12/28/2007] [Accepted: 01/18/2008] [Indexed: 02/10/2023] Open
Abstract
Background Rheumatoid arthritis (RA) is a chronic, inflammatory and systemic autoimmune disease that leads to progressive cartilage destruction. Advances in the treatment of RA-related destruction of cartilage require profound insights into the molecular mechanisms involved in cartilage degradation. Until now, comprehensive data about the molecular RA-related dysfunction of chondrocytes have been limited. Hence, the objective of this study was to establish a standardized in vitro model to profile the key regulatory molecules of RA-related destruction of cartilage that are expressed by human chondrocytes. Methods Human chondrocytes were cultured three-dimensionally for 14 days in alginate beads and subsequently stimulated for 48 hours with supernatants from SV40 T-antigen immortalized human synovial fibroblasts (SF) derived from a normal donor (NDSF) and from a patient with RA (RASF), respectively. To identify RA-related factors released from SF, supernatants of RASF and NDSF were analyzed with antibody-based protein membrane arrays. Stimulated cartilage-like cultures were used for subsequent gene expression profiling with oligonucleotide microarrays. Affymetrix GeneChip Operating Software and Robust Multi-array Analysis (RMA) were used to identify differentially expressed genes. Expression of selected genes was verified by real-time RT-PCR. Results Antibody-based protein membrane arrays of synovial fibroblast supernatants identified RA-related soluble mediators (IL-6, CCL2, CXCL1–3, CXCL8) released from RASF. Genome-wide microarray analysis of RASF-stimulated chondrocytes disclosed a distinct expression profile related to cartilage destruction involving marker genes of inflammation (adenosine A2A receptor, cyclooxygenase-2), the NF-κB signaling pathway (toll-like receptor 2, spermine synthase, receptor-interacting serine-threonine kinase 2), cytokines/chemokines and receptors (CXCL1–3, CXCL8, CCL20, CXCR4, IL-1β, IL-6), cartilage degradation (matrix metalloproteinase (MMP)-10, MMP-12) and suppressed matrix synthesis (cartilage oligomeric matrix protein, chondroitin sulfate proteoglycan 2). Conclusion Differential transcriptome profiling of stimulated human chondrocytes revealed a disturbed catabolic–anabolic homeostasis of chondrocyte function and disclosed relevant pharmacological target genes of cartilage destruction. This study provides comprehensive insight into molecular regulatory processes induced in human chondrocytes during RA-related destruction of cartilage. The established model may serve as a human in vitro disease model of RA-related destruction of cartilage and may help to elucidate the molecular effects of anti-rheumatic drugs on human chondrocyte gene expression.
Collapse
Affiliation(s)
- Kristin Andreas
- Tissue Engineering Laboratory and Berlin - Brandenburg Center for Regenerative Therapies, Department of Rheumatology, Charité - Universitätsmedizin Berlin, Tucholskystrasse 2, 10117 Berlin, Germany.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bao S, Wu Q, Sathornsumetee S, Hao Y, Li Z, Hjelmeland AB, Shi Q, McLendon RE, Bigner DD, Rich JN. Stem cell-like glioma cells promote tumor angiogenesis through vascular endothelial growth factor. Cancer Res 2007; 66:7843-8. [PMID: 16912155 DOI: 10.1158/0008-5472.can-06-1010] [Citation(s) in RCA: 964] [Impact Index Per Article: 53.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Malignant gliomas are highly lethal cancers dependent on angiogenesis. Critical tumor subpopulations within gliomas share characteristics with neural stem cells. We examined the potential of stem cell-like glioma cells (SCLGC) to support tumor angiogenesis. SCLGC isolated from human glioblastoma biopsy specimens and xenografts potently generated tumors when implanted into the brains of immunocompromised mice, whereas non-SCLGC tumor cells isolated from only a few tumors formed secondary tumors when xenotransplanted. Tumors derived from SCLGC were morphologically distinguishable from non-SCLGC tumor populations by widespread tumor angiogenesis, necrosis, and hemorrhage. To determine a potential molecular mechanism for SCLGC in angiogenesis, we measured the expression of a panel of angiogenic factors secreted by SCLGC. In comparison with matched non-SCLGC populations, SCLGC consistently secreted markedly elevated levels of vascular endothelial growth factor (VEGF), which were further induced by hypoxia. In an in vitro model of angiogenesis, SCLGC-conditioned medium significantly increased endothelial cell migration and tube formation compared with non-SCLGC tumor cell-conditioned medium. The proangiogenic effects of glioma SCLGC on endothelial cells were specifically abolished by the anti-VEGF neutralizing antibody bevacizumab, which is in clinical use for cancer therapy. Furthermore, bevacizumab displayed potent antiangiogenic efficacy in vivo and suppressed growth of xenografts derived from SCLGC but limited efficacy against xenografts derived from a matched non-SCLGC population. Together these data indicate that stem cell-like tumor cells can be a crucial source of key angiogenic factors in cancers and that targeting proangiogenic factors from stem cell-like tumor populations may be critical for patient therapy.
Collapse
Affiliation(s)
- Shideng Bao
- Department of Surgery, Preston Robert Tisch Brain Tumor Center, Molecular Cancer Biology Program, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Proost P, Mortier A, Loos T, Vandercappellen J, Gouwy M, Ronsse I, Schutyser E, Put W, Parmentier M, Struyf S, Van Damme J. Proteolytic processing of CXCL11 by CD13/aminopeptidase N impairs CXCR3 and CXCR7 binding and signaling and reduces lymphocyte and endothelial cell migration. Blood 2007; 110:37-44. [PMID: 17363734 DOI: 10.1182/blood-2006-10-049072] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
CXCR3 ligands were secreted by tissue fibroblasts and peripheral blood-derived mononuclear leukocytes in response to interferon-gamma (IFN-gamma) and Toll-like receptor (TLR) ligands. Subsequent purification and identification revealed the presence of truncated CXCL11 variants missing up to 6 amino acids. In combination with CD26/dipeptidyl peptidase IV, the metalloprotease aminopeptidase N (APN), identical to the myeloid cell marker CD13, rapidly processed CXCL11, but not CXCL8, to generate truncated CXCL11 forms. Truncated CXCL11 had reduced binding, signaling, and chemotactic properties for lymphocytes and CXCR3- or CXCR7-transfected cells. CD13/APN-truncated CXCL11 failed to induce an intracellular calcium increase but was still able to bind and desensitize CXCR3 for intact CXCL11 signaling. CXCL11 efficiently bound to CXCR7, but CXCL11 was not able to induce calcium signaling or ERK1/2 or Akt phosphorylation through CXCR7. CD26-truncated CXCL11 failed to attract lymphocytes but still inhibited microvascular endothelial cell (HMVEC) migration. However, further processing of CXCL11 by CD13 resulted in significant reduction of inhibition of HMVEC migration. Taken together, during inflammation or cancer, CXCL11 processing by CD13 may lead to a reduced number of tumor-infiltrating lymphocytes and in a more angiogenic environment.
Collapse
Affiliation(s)
- Paul Proost
- Laboratory of Molecular Immunology, Rega Institute, University of Leuven, Leuven, Belgium.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Tumor cells are known to adapt to and utilize existing physiological mechanisms to promote survival and metastasis. The role of the microenvironment in the establishment of a metastatic lesion has become increasingly important as several factors secreted by stromal cells regulate metastatic pattern in a variety of tumor types. Tumor cells interact with osteoblasts, osteoclasts and bone matrix to form a vicious cycle that is essential for successful metastases. Here we review the current concepts regarding the role of an important chemokine/chemokine receptor (SDF-1 or CXCL12/CXCR4) pathway in tumor development and metastasis. CXCL12 secretion by stromal cells is known to attract cancer cells via stimulation of the CXCR4 receptor that is up regulated by tumor cells. CXCL12/CXCR4 activation regulates the pattern of metastatic spread with organs expressing high levels of CXCL12 developing secondary tumors (i.e., the bone marrow compartment). CXCL12 has a wide range of effects in regards to tumor development but the primary role of CXCL12 appears to be the mobilization of hematopoietic stem cells and the establishment of the cancer stem-like cell niche where high levels of CXCL12 recruit a highly tumorigenic population of tumor cells and promotes cell survival, proliferation, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Jianhua Wang
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, 1011 North University Avenue, Ann Arbor, MI 48109-1078, USA
| | | | | |
Collapse
|
38
|
Tan Y, Du J, Cai S, Li X, Ma W, Guo Z, Chen H, Huang Z, Xiao J, Cai L, Cai S. Cloning and characterizing mutated human stromal cell-derived factor-1 (SDF-1): C-terminal alpha-helix of SDF-1alpha plays a critical role in CXCR4 activation and signaling, but not in CXCR4 binding affinity. Exp Hematol 2006; 34:1553-1562. [PMID: 17046575 DOI: 10.1016/j.exphem.2006.07.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2006] [Revised: 06/28/2006] [Accepted: 07/10/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE A novel C-terminal alpha-helix-defective mutant of human stromal cell-derived factor-1 (SDF-1), hSDF-154, was designed and produced in order to develop an optimal CXC chemokine receptor 4 (CXCR4) antagonist. MATERIALS AND METHODS Human native SDF-1 and alpha-helix defective SDF-1 (hSDF-154) were cloned from human bone marrow stromal cells by reverse transcription polymerase chain reaction, inserted into vector pET-30a(+), and transformed into Escherichia coli strain BL21(DE3). The recombinant hSDF-154 was purified and refolded under optimized conditions and its functional characteristics were compared with the native form of SDF-1. Functional evaluation includes migration of Jurkat and MOLT4 cells assessed by chemotaxis assay, intracellular calcium influx in these cells measured by flow cytometry, extracellular signal-regulated kinase (ERK) phosphorylation analyzed by Western blot assay, receptor binding affinity examined by sequential concentrations of unlabeled SDF-1alpha, hSDF-154 competition with (125)I- SDF-1alpha, and internalization of CXCR4 on the cell surface detected by flow cytometry. RESULTS hSDF-154 had significantly decreased chemotaxic ability, such as cell migration, as compared to the native hSDF-1. hSDF-154 failed to trigger CXCR4 to induce transient calcium influx and ERK phosphorylation. However, both hSDF-154 and the native hSDF-1 have similar binding affinity to CXCR4 and a similar ability to induce CXCR4 internalization. CONCLUSION These results indicate that hSDF-154, which has a defective C-terminal alpha-helix, a normal N-terminus, and a normal central beta-strand scaffold structure, retains normal binding affinity to CXCR4 and normal induction of CXCR4 internalization, but fails to activate CXCR4-mediated cellular signaling and chemotaxis. Therefore, the C-terminal alpha-helix of hSDF-1 plays a critical role for CXCR4 stimulation. The hSDF-154, which efficiently binds to and induces internalization of CXCR4 without activating CXCR4-related intracellular signaling and cell migration, may serve as an optimal CXCR4 antagonist.
Collapse
Affiliation(s)
- Yi Tan
- Department of Clinical Pharmacology, Pharmacy School of Jinan University, Guangzhou, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|