1
|
Ren H, Zhao Q, Wang N, Yuan X, Song R, Wen Q, Zhao Y. Melanoma-inhibiting activity promotes the migration and odontoblastic differentiation of stem cells of apical papilla. Arch Oral Biol 2025; 169:106109. [PMID: 39427554 DOI: 10.1016/j.archoralbio.2024.106109] [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: 07/21/2024] [Revised: 10/11/2024] [Accepted: 10/13/2024] [Indexed: 10/22/2024]
Abstract
OBJECTIVE Melanoma Inhibitory Activity (MIA) has been predominantly studied in the context of melanoma and cartilage development. However, its role in dental pulp development and stem cell behavior remains largely unexplored. This study investigates the expression pattern of MIA in dental pulp tissues and its potential role in the proliferation, migration, and odontoblastic differentiation of stem cells from the apical papilla (SCAPs). DESIGN MIA expression in human pulp tissue was demonstrated by immunohistochemistry. SCAPs were cultured in normal and mineralization induction media, with MIA levels monitored via RT-qPCR and Western blot. Cell proliferation was evaluated using the CCK8 assay, while transwell and cell scratch assays were conducted to examine cell migration. The effect of MIA on odontoblastic differentiation was examined by qRT-PCR, Alkaline phosphatase activity assay, and Western blot. siRNA was used to knock down MIA to investigate its effect. A mouse subcutaneous implantation model was used to assess whether MIA promotes odontoblastic differentiation in vivo. RESULTS MIA expression was observed in the papilla and odontoblasts layer of the developing pulp. In vitro, MIA expression increased during SCAPs differentiation and was found to significantly enhance migration, and odontoblastic differentiation but not proliferation. Gene knockdown experiments confirmed MIA's pivotal role in promoting SCAPs migration and differentiation. In vivo, MIA facilitated the formation of dentin-like structures and enhanced pulp-dentin complex regeneration. CONCLUSION MIA plays a crucial role in SCAPs' migration and differentiation, suggesting its potential application in pulp-dentin regeneration therapies. Further studies are required to fully elucidate the underlying mechanisms.
Collapse
Affiliation(s)
- Huihui Ren
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Qingxuan Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Nan Wang
- Department of Stomatology, Peking University Third Hospital, Beijing 100191, PR China
| | - Xiaojing Yuan
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Rui Song
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China
| | - Quan Wen
- First Clinical Division, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, 37A Xishiku Street, Xicheng District, Beijing 100034, PR China.
| | - Yuming Zhao
- Department of Pediatric Dentistry, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, No.22, Zhongguancun South Avenue, Haidian District, Beijing 100081, PR China.
| |
Collapse
|
2
|
Cao S, Wei Y, Xiong A, Yue Y, Yang J, Wang D, Liu X, Zeng H, Shi D, Li Y. Paeonol inhibits ACSL4 to protect chondrocytes from ferroptosis and ameliorates osteoarthritis progression. J Orthop Translat 2025; 50:1-13. [PMID: 39659898 PMCID: PMC11626474 DOI: 10.1016/j.jot.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/09/2024] [Accepted: 10/19/2024] [Indexed: 12/12/2024] Open
Abstract
Background Discovering an inhibitor for acyl-CoA synthetase long-chain family member 4 (ACSL4), a protein that triggers cell injury via ferroptosis, presents potential to minimize cellular damage. This study investigates paeonol (PAE), a traditional Chinese herbal medicine, as an ACSL4 inhibitor to prevent chondrocyte ferroptosis and protect against osteoarthritis (OA). Methods We conducted in vitro experiments using mouse chondrocytes treated with PAE to mitigate ferroptosis induced by Interleukin-1 Beta (IL-1β) or ferric ammonium citrate (FAC), examining intracellular ferroptotic indicators, cartilage catabolic markers, and ferroptosis regulatory proteins. A mouse OA model was created via destabilized medial meniscus (DMM), followed by intra-articular PAE injections. After 8 weeks, micro-computed tomography and histological assessments evaluated PAE's protective and anti-ferroptotic effects in the OA model. Results In vitro results showed PAE significantly reduced IL-1β/FAC-induced damage by targeting ACSL4, including cell apoptosis, inflammatory responses, extracellular matrix degradation, and ferroptotic markers (oxidative stress, lipid peroxidation, and iron buildup). It also restored the expression of ferroptotic suppressors and mitigated mitochondrial damage. Additionally, PAE increased cartilage anabolic marker expression while reducing cartilage catabolic marker expression. Molecular docking, cellular thermal shift assay, and drug affinity responsive target stability analysis verified the binding interaction between PAE and ACSL4. Furthermore, the role of PAE in chondrocytes was further verified through ACSL4 knockdown and overexpression. In vivo, mice with OA showed increased cartilage degradation and ferroptosis, while intra-articular PAE injection alleviated these pathological changes. Conclusion PAE significantly protects chondrocytes from ferroptosis induced by IL-1β/FAC in primary mouse chondrocytes and DMM surgery-induced OA mice through ACSL4 inhibition. The translational potential of this article These findings highlight the potential of targeting ACSL4 in chondrocytes as a treatment strategy for OA, positioning PAE as a promising drug candidate.
Collapse
Affiliation(s)
- Siyang Cao
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
- Faculty of Pharmaceutical Sciences, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences (CAS), Shenzhen, Guangdong, China
| | - Ao Xiong
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yaohang Yue
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jun Yang
- Department of Radiology, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Deli Wang
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone & Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Xiyu Liu
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Hui Zeng
- National & Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Orthopedics, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, China
| | - Dongquan Shi
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopaedic Surgery, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Ye Li
- Department of Rehabilitation Science, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region, China
| |
Collapse
|
3
|
Staebler S, Lichtblau A, Gurbiel S, Schubert T, Riechers A, Rottensteiner-Brandl U, Bosserhoff A. MIA/CD-RAP Regulates MMP13 and Is a Potential New Disease-Modifying Target for Osteoarthritis Therapy. Cells 2023; 12:cells12020229. [PMID: 36672165 PMCID: PMC9856983 DOI: 10.3390/cells12020229] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/02/2023] [Accepted: 01/04/2023] [Indexed: 01/06/2023] Open
Abstract
Melanoma inhibitory activity/cartilage-derived retinoicacid-sensitive protein (MIA/CD-RAP) is a protein expressed and secreted by chondrocytes and cartilaginous tissues. MIA/CD-RAP-deficient mice develop milder osteoarthritis than wildtype mice. In this study, we investigated MIA/CD-RAP downstream targets to explain this reduced disease development. As a possible mediator, we could detect matrix metalloproteinase 13 (MMP13), and the influence of MIA/CD-RAP on MMP13 regulation was analyzed in vitro using SW1353 chondrosarcoma cells and primary chondrocytes. The femoral head cartilage of WT and MIA/CD-RAP -/- mice were cultured ex vivo to further investigate MMP13 activity. Finally, osteoarthritis was surgically induced via DMM in C57BL/6 mice, and the animals were treated with an MIA/CD-RAP inhibitory peptide by subcutaneously implanted pellets. MMP13 was regulated by MIA/CD-RAP in SW1353 cells, and MIA/CD-RAP -/- murine chondrocytes showed less expression of MMP13. Further, IL-1β-treated MIA/CD-RAP -/- chondrocytes displayed less MMP13 expression and activity. Additionally, MIA/CD-RAP-deficient ex vivo cultured cartilage explants showed less MMP13 activity as well as reduced cartilage degradation. The mice treated with the MIA/CD-RAP inhibitory peptide showed less osteoarthritis development. Our findings revealed MIA/CD-RAP as a new regulator of MMP13 and highlighted its role as a potential new target for osteoarthritis therapy.
Collapse
Affiliation(s)
- Sebastian Staebler
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Adrian Lichtblau
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Slavyana Gurbiel
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
| | - Thomas Schubert
- Institute of Pathology, Friedrich-Alexander Universität Erlangen-Nürnberg (FAU), Krankenhausstraße 8-10, 91054 Erlangen, Germany
- Institute of Applied Pathology, 67346 Speyer, Germany
| | - Alexander Riechers
- Institute of Pathology, Medical School, University of Regensburg, 93053 Regensburg, Germany
| | | | - Anja Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91052 Erlangen, Germany
- Correspondence:
| |
Collapse
|
4
|
Bian Q, Cheng YH, Wilson JP, Su EY, Kim DW, Wang H, Yoo S, Blackshaw S, Cahan P. A single cell transcriptional atlas of early synovial joint development. Development 2020; 147:dev185777. [PMID: 32580935 PMCID: PMC7390639 DOI: 10.1242/dev.185777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 06/09/2020] [Indexed: 12/14/2022]
Abstract
Synovial joint development begins with the formation of the interzone, a region of condensed mesenchymal cells at the site of the prospective joint. Recently, lineage-tracing strategies have revealed that Gdf5-lineage cells native to and from outside the interzone contribute to most, if not all, of the major joint components. However, there is limited knowledge of the specific transcriptional and signaling programs that regulate interzone formation and fate diversification of synovial joint constituents. To address this, we have performed single cell RNA-Seq analysis of 7329 synovial joint progenitor cells from the developing murine knee joint from E12.5 to E15.5. By using a combination of computational analytics, in situ hybridization and in vitro characterization of prospectively isolated populations, we have identified the transcriptional profiles of the major developmental paths for joint progenitors. Our freely available single cell transcriptional atlas will serve as a resource for the community to uncover transcriptional programs and cell interactions that regulate synovial joint development.
Collapse
Affiliation(s)
- Qin Bian
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Yu-Hao Cheng
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Jordan P Wilson
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Emily Y Su
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Dong Won Kim
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Hong Wang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Sooyeon Yoo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Seth Blackshaw
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| | - Patrick Cahan
- Institute for Cell Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Biomedical Engineering, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
- Department of Molecular Biology and Genetics, Johns Hopkins School of Medicine, Baltimore MD 21205, USA
| |
Collapse
|
5
|
ER-to-Golgi Transport: A Sizeable Problem. Trends Cell Biol 2019; 29:940-953. [DOI: 10.1016/j.tcb.2019.08.007] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 08/22/2019] [Accepted: 08/23/2019] [Indexed: 11/16/2022]
|
6
|
Feuerer L, Lamm S, Henz I, Kappelmann-Fenzl M, Haferkamp S, Meierjohann S, Hellerbrand C, Kuphal S, Bosserhoff AK. Role of melanoma inhibitory activity in melanocyte senescence. Pigment Cell Melanoma Res 2019; 32:777-791. [PMID: 31172672 DOI: 10.1111/pcmr.12801] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 05/16/2019] [Accepted: 05/26/2019] [Indexed: 01/10/2023]
Abstract
The protein melanoma inhibitory activity (MIA) is known to be expressed in melanoma and to support melanoma progression. Interestingly, previous studies also observed the expression of MIA in nevi. Concentrating on these findings, we revealed that MIA expression is correlated with a senescent state in melanocytes. Induction of replicative or oncogene-induced senescence resulted in increased MIA expression in vitro. Notably, MIA knockdown in senescent melanocytes reduced the percentage of senescence-associated beta-Gal-positive cells and enhanced proliferation. Using the melanoma mouse model Tg(Grm1), MIA-deficient mice supported the impact of MIA on senescence by showing a significantly earlier tumor onset compared to controls. In melanocytes, MIA knockdown led to a downregulation of the cell cycle inhibitor p21 in vitro and in vivo. In contrast, after induction of hTERT in human melanoma cells, p21 regulation by MIA was lost. In summary, our data show for the first time that MIA is a regulator of cellular senescence in human and murine melanocytes.
Collapse
Affiliation(s)
- Lena Feuerer
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Lamm
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Ingmar Henz
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Melanie Kappelmann-Fenzl
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany.,Deggendorf Institute of Technology, Deggendorf, Germany
| | - Sebastian Haferkamp
- Department of Dermatology, University Hospital Regensburg, Regensburg, Germany
| | | | - Claus Hellerbrand
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| | - Silke Kuphal
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Anja Katrin Bosserhoff
- Emil Fischer Center, Institute of Biochemistry, University of Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| |
Collapse
|
7
|
Hypoxia-Inducible Factor 1 Is an Inductor of Transcription Factor Activating Protein 2 Epsilon Expression during Chondrogenic Differentiation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:380590. [PMID: 26273614 PMCID: PMC4530219 DOI: 10.1155/2015/380590] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Revised: 07/03/2015] [Accepted: 07/06/2015] [Indexed: 02/07/2023]
Abstract
The transcription factor AP-2ε (activating enhancer-binding protein epsilon) is expressed in cartilage of humans and mice. However, knowledge about regulatory mechanisms influencing AP-2ε expression is limited. Using quantitative real time PCR, we detected a significant increase in AP-2ε mRNA expression comparing initial and late stages of chondrogenic differentiation processes in vitro and in vivo. Interestingly, in these samples the expression pattern of the prominent hypoxia marker gene angiopoietin-like 4 (Angptl4) strongly correlated with that of AP-2ε suggesting that hypoxia might represent an external regulator of AP-2ε expression in mammals. In order to show this, experiments directly targeting the activity of hypoxia-inducible factor-1 (HIF1), the complex mediating responses to oxygen deprivation, were performed. While the HIF1-activating compounds 2,2'-dipyridyl and desferrioxamine resulted in significantly enhanced mRNA concentration of AP-2ε, siRNA against HIF1α led to a significantly reduced expression rate of AP-2ε. Additionally, we detected a significant upregulation of the AP-2ε mRNA level after oxygen deprivation. In sum, these different experimental approaches revealed a novel role for the HIF1 complex in the regulation of the AP-2ε gene in cartilaginous cells and underlined the important role of hypoxia as an important external regulatory stimulus during chondrogenic differentiation modulating the expression of downstream transcription factors.
Collapse
|
8
|
Niebler S, Schubert T, Hunziker EB, Bosserhoff AK. Activating enhancer binding protein 2 epsilon (AP-2ε)-deficient mice exhibit increased matrix metalloproteinase 13 expression and progressive osteoarthritis development. Arthritis Res Ther 2015; 17:119. [PMID: 25964075 PMCID: PMC4453098 DOI: 10.1186/s13075-015-0648-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 05/05/2015] [Indexed: 01/15/2023] Open
Abstract
Introduction The transcription factor activating enhancer binding protein 2 epsilon (AP-2ε) was recently shown to be expressed during chondrogenesis as well as in articular chondrocytes of humans and mice. Furthermore, expression of AP-2ε was found to be upregulated in affected cartilage of patients with osteoarthritis (OA). Despite these findings, adult mice deficient for AP-2ε (Tfap2e−/−) do not exhibit an obviously abnormal cartilaginous phenotype. We therefore analyzed embryogenesis of Tfap2e−/− mice to elucidate potential transient abnormalities that provide information on the influence of AP-2ε on skeletal development. In a second part, we aimed to define potential influences of AP-2ε on articular cartilage function and gene expression, as well as on OA progression, in adult mice. Methods Murine embryonic development was accessed via in situ hybridization, measurement of skeletal parameters and micromass differentiation of mesenchymal cells. To reveal discrepancies in articular cartilage of adult wild-type (WT) and Tfap2e−/− mice, light and electron microscopy, in vitro culture of cartilage explants, and quantification of gene expression via real-time PCR were performed. OA was induced via surgical destabilization of the medial meniscus in both genotypes, and disease progression was monitored on histological and molecular levels. Results Only minor differences between WT and embryos deficient for AP-2ε were observed, suggesting that redundancy mechanisms effectively compensate for the loss of AP-2ε during skeletal development. Surprisingly, though, we found matrix metalloproteinase 13 (Mmp13), a major mediator of cartilage destruction, to be significantly upregulated in articular cartilage of adult Tfap2e−/− mice. This finding was further confirmed by increased Mmp13 activity and extracellular matrix degradation in Tfap2e−/− cartilage explants. OA progression was significantly enhanced in the Tfap2e−/− mice, which provided evidence for in vivo relevance. This finding is most likely attributable to the increased basal Mmp13 expression level in Tfap2e−/− articular chondrocytes that results in a significantly higher total Mmp13 expression rate during OA as compared with the WT. Conclusions We reveal a novel role of AP-2ε in the regulation of gene expression in articular chondrocytes, as well as in OA development, through modulation of Mmp13 expression and activity.
Collapse
Affiliation(s)
- Stephan Niebler
- Institute of Biochemistry (Emil-Fischer-Center), Friedrich Alexander University Erlangen-Nürnberg, Fahrstrasse17, 91054, Erlangen, Germany. .,Institute of Pathology, University Regensburg, Franz-Josef-Strauss-Allee 11, 93053, Regensburg, Germany.
| | - Thomas Schubert
- Institute of Pathology, Friedrich Alexander University Erlangen-Nürnberg, Krankenhausstrasse 8-10, 91054, Erlangen, Germany.
| | - Ernst B Hunziker
- Department of Orthopedic Surgery, University Hospital of Bern, Murtenstrasse 35, 3010, Bern, Switzerland.
| | - Anja K Bosserhoff
- Institute of Biochemistry (Emil-Fischer-Center), Friedrich Alexander University Erlangen-Nürnberg, Fahrstrasse17, 91054, Erlangen, Germany.
| |
Collapse
|
9
|
Docheva D, Popov C, Alberton P, Aszodi A. Integrin signaling in skeletal development and function. ACTA ACUST UNITED AC 2014; 102:13-36. [DOI: 10.1002/bdrc.21059] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Accepted: 01/14/2014] [Indexed: 12/22/2022]
Affiliation(s)
- Denitsa Docheva
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| | - Cvetan Popov
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| | - Paolo Alberton
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| | - Attila Aszodi
- Experimental Surgery and Regenerative Medicine, Department of Surgery; Ludwig-Maximilians-University; 80336 Munich Germany
| |
Collapse
|
10
|
Graf SA, Busch C, Bosserhoff AK, Besch R, Berking C. SOX10 promotes melanoma cell invasion by regulating melanoma inhibitory activity. J Invest Dermatol 2014; 134:2212-2220. [PMID: 24608986 DOI: 10.1038/jid.2014.128] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 01/26/2014] [Accepted: 02/14/2014] [Indexed: 12/23/2022]
Abstract
The transcription factor SOX10 (SRY (sex determining region Y)-box 10) has a key role in the embryonic development of melanocytes. Recently, it has been suggested that SOX10 is highly relevant for melanoma development and survival. However, the distinct functions and downstream targets of SOX10 in melanoma remain widely unknown. In this study, we inhibited SOX10 via RNA interference in different human melanoma cell lines and found a significantly reduced invasion capacity in vitro and in the chick embryo model. At later time points, SOX10 inhibition reduced proliferation and induced cell death. We identified melanoma inhibitory activity (MIA) as a direct target gene of SOX10, which is an essential protein for melanoma cell migration and invasion. Expression levels of SOX10 and MIA strictly correlated in melanoma cell lines, and SOX10 inhibition reduced MIA expression and promoter activity. Direct binding of SOX10 to the MIA promoter was demonstrated by electrophoretic mobility shift assay and chromatin immunoprecipitation. Ectopic expression of MIA in SOX10-inhibited melanoma cells restored the invasion capacity, supporting the hypothesis that MIA is responsible for SOX10-mediated melanoma cell invasion. Our data provide evidence for a critical role of SOX10 in melanoma cell invasion through the regulation of MIA and highlight its role as a therapeutic target in melanoma.
Collapse
Affiliation(s)
- Saskia A Graf
- Department of Dermatology and Allergology, Ludwig-Maximilian University, Munich, Germany
| | - Christian Busch
- Section of Dermato-Oncology, Department of Dermatology, University of Tuebingen, Tuebingen, Germany
| | | | - Robert Besch
- Department of Dermatology and Allergology, Ludwig-Maximilian University, Munich, Germany
| | - Carola Berking
- Department of Dermatology and Allergology, Ludwig-Maximilian University, Munich, Germany.
| |
Collapse
|
11
|
Abstract
Osteoarthritis (OA) is a slowly progressing, degenerative disorder of synovial joints culminating in the irreversible destruction of articular cartilage and subchondral bone. It affects almost everyone over the age of 65 and influences life quality of affected individuals with enormous costs to the health care system. Current therapeutic strategies seek to ameliorate pain and increase mobility; however, to date none of them halts disease progression or regenerates damaged cartilage or bone. Thus, there is an ultimate need for the development of new, noninvasive treatments that could substitute joint replacement for late- or end-stage patients. Therefore, osteoarthritis animal models for mimicking of all OA features are important. Mice develop an OA pathology that is comparable to humans, rapidly develop OA due to the short lifetime and show reproducible OA symptoms. They provide a versatile and widely used animal model for analyzing molecular mechanisms of OA pathology. One major advantage over large animal models is the availability of knockout or transgenic mice strains to examine genetic predispositions/contributions to OA.In this chapter, we describe three widely used instability-inducing murine osteoarthritis models. The most common two methods for surgical induction are: (1) destabilization of the medial meniscus (DMM) and (2) anterior cruciate ligament transection (ACLT). In the DMM model, the medial meniscotibial ligament is transected while in the ACLT model the anterior cruciate ligament is destroyed. In the third, chemical induced instability method, intraarticular collagenase is injected into the knee joint. Intraarticular collagenase weakens articular ligaments which cause instability of the joint, and full-blown OA develops within 6 weeks. For morphological evaluation, we correspond mainly to the recommendations of OARSI for histological assessment of osteoarthritis in mouse. For statistical evaluation summed or mean scores of all four knee areas (medial tibial plateau (MTP), medial tibial condyle (MFC), lateral tibial plateau (LTP) or lateral femoral condyle (LFC)), medial and/or lateral regions are used.In future, not only large animal models like guinea pigs, sheep, goats, or horses will be important for a better understanding of osteoarthritis, but especially the mouse model with its rapid development of osteoarthritis and its numerous advantages by providing knockout or transgenic strains will become more and more relevant for drug development and determination of genetic predispositions of osteoarthritis pathology.
Collapse
|
12
|
Schmid R, Meyer K, Spang R, Schittek B, Bosserhoff AK. YBX1 is a modulator of MIA/CD-RAP-dependent chondrogenesis. PLoS One 2013; 8:e82166. [PMID: 24349210 PMCID: PMC3861381 DOI: 10.1371/journal.pone.0082166] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2013] [Accepted: 10/31/2013] [Indexed: 11/19/2022] Open
Abstract
MIA/CD-RAP is a small, secreted protein involved in cartilage differentiation and melanoma progression. We recently revealed that p54(nrb) acts as a mediator of MIA/CD-RAP action to promote chondrogenesis and the progression of malignant melanoma. As the molecular mechanism of MIA/CD-RAP action in cartilage has not been defined in detail until now, we aimed to understand the regulation of p54(nrb) transcription in chondrogenesis. We concentrated on the previously described MIA/CD-RAP-dependent regulatory region in the p54(nrb) promoter and characterized the transcriptional regulation of p54(nrb) by MIA/CD-RAP in cartilage. A series of truncated p54(nrb) promoter constructs and mutagenesis analysis revealed that the transcription factor YBX1, which has not been investigated in chondrogenesis thus far, is the mediator of MIA/CD-RAP dependent activation of p54(nrb) transcription. A systematic analysis of genes carrying this binding site in their promoter region revealed further potential MIA/CD-RAP-regulated genes that have been implicated in cartilage differentiation. In summary, we described the effects of MIA/CD-RAP on transcriptional regulation in chondrocytes. Understanding the regulation of p54(nrb) via YBX1 contributes to the understanding of chondrogenesis. Uncovering new downstream effectors that function via the activation of YBX1 supports the important role of MIA/CD-RAP in these processes.
Collapse
Affiliation(s)
- Rainer Schmid
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | - Katharina Meyer
- Institute for Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Rainer Spang
- Institute for Functional Genomics, University of Regensburg, Regensburg, Germany
| | - Birgit Schittek
- Institute of Dermatology, University of Tübingen, Tübingen, Germany
| | | |
Collapse
|
13
|
Yeremenko N, Härle P, Cantaert T, van Tok M, van Duivenvoorde LM, Bosserhoff A, Baeten D. The cartilage protein melanoma inhibitory activity contributes to inflammatory arthritis. Rheumatology (Oxford) 2013; 53:438-47. [PMID: 24287514 DOI: 10.1093/rheumatology/ket382] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVE Melanoma inhibitory activity (MIA) is a small chondrocyte-specific protein with unknown function. MIA knockout mice (MIA(-/-)) have a normal phenotype with minor microarchitectural alterations of cartilage. Our previous study demonstrated that immunodominant epitopes of MIA are actively presented in an HLA-DR4-restricted manner in the inflamed RA joint. The objective of this study was to investigate the potential role of MIA as an autoantigen. METHODS Collagen-induced arthritis (CIA) and anti-collagen antibody-induced arthritis (CAIA) were induced in MIA(-/-) mice. Anti-type II collagen (anti-CII) antibodies were measured by ELISA. T cell proliferation and cytokine production were assessed by flow cytometry. RESULTS MIA(-/-) mice had a markedly reduced incidence and severity of CIA and CAIA compared with wild-type (WT) mice. Attenuation of disease was not related to defective binding of anti-CII antibodies to cartilage in the absence of MIA. However, MIA(-/-) mice had significantly reduced anti-CII IgG1 and IgG2a antibody levels accompanied by an increase in FoxP3-expressing CD25(+)CD4(+) regulatory T cells. This was paralleled by a significant reduction in CII-specific IFN-γ production by T cells in MIA(-/-) but not WT animals, suggesting a qualitative impact of MIA on the collagen-induced Th1 response. Furthermore, Ag-specific proliferation of T cells after restimulation with MIA in WT but not MIA(-/-) mice indicated the existence of MIA-specific T cells in the context of CIA. CONCLUSION These data support a role for MIA as an autoantigen during arthritis development. Whether MIA can influence the balance of pathogenic vs regulatory responses in human RA remains to be investigated.
Collapse
Affiliation(s)
- Nataliya Yeremenko
- Department of Clinical Immunology and Rheumatology, Academic Medical Center/University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
14
|
Schmid R, Bosserhoff AK. Redundancy in regulation of chondrogenesis in MIA/CD-RAP-deficient mice. Mech Dev 2013; 131:24-34. [PMID: 24269712 DOI: 10.1016/j.mod.2013.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Revised: 10/26/2013] [Accepted: 11/11/2013] [Indexed: 10/26/2022]
Abstract
Recent in vitro analysis of MIA/CD-RAP-deficient (MIA(-/-)) mesenchymal stem cells revealed altered chondrogenic differentiation, characterised by enhanced proliferation and delayed differentiation. However, adult MIA(-/-) mice develop normally and show only ultrastructural defects of the cartilage but no major abnormalities. We therefore focused, in this study, on chondrogenesis in vivo in MIA(-/-) mouse embryos to reveal potential molecular changes during embryogenesis and possible redundant mechanisms, which explain the almost normal phenotype despite MIA/CD-RAP loss. In situ hybridisation analysis revealed larger expression areas of Col2a1 and Sox9 positive, proliferating chondrocytes at day 15.5 and 16.5 of embryogenesis in MIA(-/-) mice. The initially diminished zone of Col10a1-expressing hypertrophic chondrocytes at day 15.5 was compensated at day 16.5 in MIA(-/-) embryos. Supported by in vitro studies using mesenchymal stem cells, we discovered that chondrogenesis in MIA(-/-) mice is modified by enhanced Sox9, Sox6 and AP-2α expression. Finally, we identified reduced AP1 and CRE activity, analysed by reporter gene- and electrophoretic mobility shift assays, important for redundancy mechanism which rescued delayed hypertrophic differentiation and allows normal development of MIA(-/-) mice. In summary, as observed in other knockout models of molecules important for cartilage development and differentiation, viability and functional integrity is reached by remarkable molecular redundancy in MIA/CD-RAP knockout mice.
Collapse
Affiliation(s)
- Rainer Schmid
- University of Regensburg Medical School, Institute of Pathology, D-93053 Regensburg, Germany
| | - Anja-Katrin Bosserhoff
- University of Regensburg Medical School, Institute of Pathology, D-93053 Regensburg, Germany.
| |
Collapse
|
15
|
Ruedel A, Hofmeister S, Bosserhoff AK. Development of a model system to analyze chondrogenic differentiation of mesenchymal stem cells. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2013; 6:3042-3048. [PMID: 24294400 PMCID: PMC3843294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 09/17/2013] [Accepted: 10/02/2013] [Indexed: 06/02/2023]
Abstract
High-density cell culture is widely used for the analysis of cartilage development of human mesenchymal stem cells (HMSCs) in vitro. Several cell culture systems, as micromass, pellet culture and alginate culture, are applied by groups in the field to induce chondrogenic differentiation of HMSCs. A draw back of all model systems is the high amount of cells necessary for the experiments. Further, handling of large experimental approaches is difficult due to culturing e.g. in 15 ml tubes. Therefore, we aimed to develop a new model system based on "hanging drop" cultures using 10 to 100 fold less cells. Here, we demonstrate that differentiation of chondrogenic cells was induced as previously shown in other model systems. Real time RT-PCR analysis demonstrated that Collagen type II and MIA/CD-RAP were upregulated during culturing whereas for induction of hypertrophic markers like Collagen type X and AP-2 epsilon treatment with TGF beta was needed. To further test the system, siRNA against Sox9 was used and effects on chondrogenic gene expression were evaluated. In summary, the hanging drop culture system was determined to be a promising tool for in vitro chondrogenic studies.
Collapse
Affiliation(s)
- Anke Ruedel
- Institute of Pathology, University of Regensburg Medical School Franz-Josef-Strauss-Allee 11, D-93053 Regensburg, Germany
| | | | | |
Collapse
|
16
|
Zanfardino M, Spampanato C, De Cicco R, Buommino E, De Filippis A, Baiano S, Barra A, Morelli F. Simvastatin reduces melanoma progression in a murine model. Int J Oncol 2013; 43:1763-70. [PMID: 24101161 PMCID: PMC3833984 DOI: 10.3892/ijo.2013.2126] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 07/04/2013] [Indexed: 12/27/2022] Open
Abstract
Statins are a class of drugs that inhibit the rate-limiting step in the cholesterol biosynthetic pathway and show an anticancer effect, probably through the inhibition of cell proliferation. To date, the exact mechanism of cancer cell growth arrest induced by statins is not known. We report that simvastatin is able to induce apoptosis in melanoma cells but not in normal cells and also able to contrast the growth of tumor in an experimental melanoma murine model. We observed a delay in the tumor development in almost the 50% of the simvastatin administered animals and a strong reduction of the tumor volume with a differences of ~150% compared to the controls. Also the survival rate was significantly higher in mice that received the drug with a survival increase of ~130% compared to the controls. The tumor growth reduction in mice was supported by the results of cell migration assay, confirming that simvastatin clearly reduced cell migration. Moreover, simvastatin induced a strong downregulation of NonO gene expression, an important growth factor involved in the splicing regulation. This result could explain the decrease of melanoma cells proliferation, suggesting a possible action mechanism. The results derived from our experiments may sustain the many reports on the anticancer inhibitory property of statins and encourage new studies on this drug for a possible use in therapy, probably in combination with conventional chemotherapy.
Collapse
Affiliation(s)
- Mario Zanfardino
- Institute of Genetics and Biophysics A. Buzzati Traverso, CNR Naples, Naples, Italy
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Schmid R, Meyer K, Spang R, Schittek B, Bosserhoff AK. Melanoma inhibitory activity promotes melanoma development through activation of YBX1. Pigment Cell Melanoma Res 2013; 26:685-96. [PMID: 23672612 DOI: 10.1111/pcmr.12119] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2013] [Accepted: 05/08/2013] [Indexed: 02/06/2023]
Abstract
Melanoma inhibitory activity (MIA), a small soluble secreted protein, is functionally important for progression of malignant melanoma. We recently revealed that p54(nrb) acts as a mediator of MIA action. In this study, we characterize the transcriptional regulation of p54(nrb) by MIA to explain MIA's molecular action. We identified one highly conserved region in the p54(nrb) promoter that is necessary and sufficient for MIA-dependent activation. Functional promoter analysis identified the transcription factor YBX1 as the mediator of MIA activation of p54(nrb) transcription. We screened the genome for further potential MIA-regulated genes carrying the element in their promoter regions. Integrating our sequence data with expression data from human melanomas identified a list of 23 potential MIA-YBX1 targets in melanomas. In summary, we present for the first time effects of MIA on transcriptional regulation. Uncovering new potential downstream effectors working via activation of YBX1 supports the important role of MIA in melanoma.
Collapse
Affiliation(s)
- Rainer Schmid
- Institute of Pathology, University of Regensburg Medical School, Regensburg, Germany
| | | | | | | | | |
Collapse
|
18
|
Little CB, Hunter DJ. Post-traumatic osteoarthritis: from mouse models to clinical trials. Nat Rev Rheumatol 2013; 9:485-97. [PMID: 23689231 DOI: 10.1038/nrrheum.2013.72] [Citation(s) in RCA: 194] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Osteoarthritis (OA), the most common of all arthropathies, is a leading cause of disability and has a large (and growing) worldwide socioeconomic cost. Despite its burgeoning importance, translation of disease-modifying OA therapies from the laboratory into clinical practice has slowed. Differences between the OA models studied preclinically and the disease evaluated in human clinical trials contribute to this failure. Most animal models of OA induce disease through surgical or mechanical disruption of joint biomechanics in young individuals rather than the spontaneous development of age-associated disease. This instability-induced joint disease in animals best models the arthritis that develops in humans after an injurious event, known as post-traumatic OA (PTOA). Studies in genetically modified mice suggest that PTOA has a distinct molecular pathophysiology compared with that of spontaneous OA, which might explain the poor translation from preclinical to clinical OA therapeutic trials. This Review summarizes the latest data on potential molecular targets for PTOA prevention and modification derived from studies in genetically modified mice, and describes their validation in preclinical therapeutic trials. This article focuses on how these findings might best be translated to humans, and identifies the potential challenges to successful implementation of clinical trials of disease-modifying drugs for PTOA.
Collapse
Affiliation(s)
- Christopher B Little
- Raymond Purves Bone and Joint Research Laboratories, Kolling Institute of Medical Research, Institute of Bone and Joint Research, University of Sydney at Royal North Shore Hospital, St Leonards, NSW 2065, Australia. christopher.little@ sydney.edu.au
| | | |
Collapse
|
19
|
Kappelmann M, Kuphal S, Meister G, Vardimon L, Bosserhoff AK. MicroRNA miR-125b controls melanoma progression by direct regulation of c-Jun protein expression. Oncogene 2012; 32:2984-91. [PMID: 22797068 DOI: 10.1038/onc.2012.307] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
A fundamental event in the development and progression of malignant melanoma is the deregulation of cancer-relevant transcription factors. We recently showed that c-Jun is a main regulator of tumor progression in melanoma and thus the most important member of the AP-1 transcription factor family for this disease. Interestingly, we revealed that c-Jun expression was regulated on the post-transcriptional level and therefore speculated that miRNAs could be involved in c-Jun regulation. We determined seed sequences for miR-125b and miR-527 in the coding region of c-Jun mRNA that hints at the direct involvement of miRNA-dependent regulation on the protein level. We found that the expression of miR-125b was significantly reduced in malignant melanoma cell lines and tissue samples compared with melanocytes, whereas miR-527 remained unchanged. In further functional experiments, treatment of melanoma cells with pre-miR-125b resulted in strong suppression of cellular proliferation and migration, supporting the role of miR-125b in melanoma. In addition, transfection of pre-miR-125b led to strong downregulation of c-Jun protein but not mRNA expression in melanoma cells. Luciferase assays using reporter plasmids containing the miR-125b seed sequence in the luciferase coding region confirmed the direct interaction with miR-125b. Furthermore, immunoprecipitation of Ago-2 revealed that c-Jun mRNA accumulated in the RNA-induced silencing complex after pre-miR-125b transfection in melanoma cells. In summary, we identified an important role for miR-125b in malignant melanoma. Moreover, we demonstrated post-transcriptional regulation of c-Jun by this miRNA and showed that c-Jun is a main mediator of the effects of miR-125b on melanoma cells.
Collapse
Affiliation(s)
- M Kappelmann
- Institute of Pathology, University of Regensburg, Regensburg, Germany
| | | | | | | | | |
Collapse
|
20
|
Riechers A, Bosserhoff AK. Pitfalls in immunohistochemistry--a recent example. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2012; 5:137-139. [PMID: 22400073 PMCID: PMC3294226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 01/31/2012] [Accepted: 02/07/2012] [Indexed: 05/31/2023]
Abstract
Immunohistochemistry is an important and valuable technique in many fields of research, although several common pitfalls can lead to wrong or misinterpreted results. A recently published study [1] claims that the protein MIA (melanoma inhibitory activity) is expressed in Purkinje cells in the cerebellum. Careful re-analysis resulted in negative results. Due to these results of our group we feel that this analysis could serve as example for the potential problems in immunohistochemistry caused by the combination of an unspecific antibody and the omission of evaluating control tissue samples.
Collapse
|
21
|
Bucur O, Stancu AL, Khosravi-Far R, Almasan A. Analysis of apoptosis methods recently used in Cancer Research and Cell Death & Disease publications. Cell Death Dis 2012; 3:e263. [PMID: 22297295 PMCID: PMC3288344 DOI: 10.1038/cddis.2012.2] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
22
|
Albrecht C, Tichy B, Nürnberger S, Hosiner S, Zak L, Aldrian S, Marlovits S. Gene expression and cell differentiation in matrix-associated chondrocyte transplantation grafts: a comparative study. Osteoarthritis Cartilage 2011; 19:1219-27. [PMID: 21820068 DOI: 10.1016/j.joca.2011.07.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 06/29/2011] [Accepted: 07/05/2011] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Although scaffold composition and architecture are considered to be important parameters for tissue engineering, their influence on gene expression and cell differentiation is rarely investigated in scaffolds used for matrix-associated autologous chondrocyte transplantation (MACT). In this study we have therefore comparatively analyzed the gene expression of important chondrogenic markers in four clinical applied cell-graft systems with very different scaffold characteristics. METHODS Residuals (n=165) of four different transplant types (MACI®, Hyalograft®C, CaReS® and Novocart®3D) were collected during surgery and analyzed for Col1, Col2, aggrecan, versican, melanoma inhibitory activity (MIA) and IL-1β by real-time PCR. Scaffold and cell morphology were evaluated by histology and electron microscopy. RESULTS Despite the cultivation on 3D scaffolds, the cell differentiation on all transplant types didn't reach the levels of native cartilage. Gene expression highly differed between the transplant types. The highest differentiation of cells (Col2/Col1 ratio) was found in CaReS®, followed by Novocart®3D, Hyalograft®C and MACI®. IL-1β expression also exhibited high differences between the scaffolds showing low expression levels in Novocart®3D and CaReS® and higher expression levels in MACI® and Hyalograft®C. CONCLUSIONS Our data indicate that scaffold characteristics as well as culture conditions highly influence gene expression in cartilage transplants and that these parameters may have profound impact on the tissue regeneration after MACT.
Collapse
Affiliation(s)
- C Albrecht
- Medical University of Vienna, Department of Traumatology, Center for Joint and Cartilage, Vienna, Austria.
| | | | | | | | | | | | | |
Collapse
|
23
|
Abstract
The strong interest in cell death, and the shift in emphasis from basic mechanisms to translational aspects fostered the launch last year of the new sister journal of Cell Death and Differentiation, named Cell Death and Disease, to reflect its stronger focus towards clinical applications. Here, we review that first year of activity, which reflects an enthusiastic response by the scientific community. On the basis of this, we now launch two novel initiatives, the start of a new section dedicated to cancer metabolism and the opening of a new editorial office in Shanghai.
Collapse
|