1
|
Liu X, Zhang X, Chen Y, Tang J, Zhang H, Jin G. SSX2IP promotes cell proliferation and migration in breast cancer by regulating FANCI. Cell Biol Int 2025; 49:188-200. [PMID: 39533770 DOI: 10.1002/cbin.12259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 09/18/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Synovial sarcoma X breakpoint 2 interacting protein (SSX2IP) is expressed in various normal tissues and participates in the progression of human cancers. Nevertheless, the specific functions and underlying molecular mechanisms of SSX2IP in cancer, particularly in breast cancer, remain poorly understood. In this study, we aimed to explore the functional role of SSX2IP in breast cancer. Immunohistochemical staining, quantitative real-time PCR, and western blotting blot analysis were used to assess genes expression levels. By manipulating SSX2IP expression levels and conducting functional assays including Celigo cell counting assay or CCKCCK-8-8 assay, flow cytometry, wound healing assay, and Transwell assay, we explored the impact of SSX2IP on the malignant phenotype of breast cancer cells. Additionally, the in vivo tumor-suppressive ability of SSX2IP was investigated by tumor xenograft experiment. Our results revealed an upregulation of SSX2IP in the breast cancer. Functional assays demonstrated that SSX2IP knockdown inhibited cell proliferation and migration, induced apoptosis in vitro, as well as suppressed the tumor growth in vivo. Conversely, SSX2IP overexpression contributed to the malignant phenotype of breast cancer cells. Co-expression analysis showed that FA Complementation Group I (FANCI) was co-expressed with SSX2IP. Additionally, SSX2IP positively regulated FANCI expression and its interaction was verified by Co-IP.Co-IP. Furthermore, FANCI overexpression partially reversed the effects of SSX2IP knockdown on cell proliferation and metastasis. In summary, our findings revealed that SSX2IP contributes to the progression of breast cancer by regulating FANCI, hinting at its potential as a novel biomarker and therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Xianfu Liu
- Department of Surgical Oncology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xiaojing Zhang
- Department of Surgical Oncology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yansong Chen
- Department of Surgical Oncology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jingwei Tang
- Department of Surgical Oncology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Hao Zhang
- Department of Surgical Oncology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Gongsheng Jin
- Department of Surgical Oncology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
2
|
Fu X, He Y, Song J, Wang L, Guo P, Cao J. MiRNA-181b-5p Modulates Cell Proliferation, Cell Cycle and Apoptosis by Targeting SSX2IP in Acute Lymphoblastic Leukemia. Turk J Haematol 2022; 39:160-169. [PMID: 35658330 PMCID: PMC9421343 DOI: 10.4274/tjh.galenos.2022.2022.0054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Objective: Accumulating evidence indicates that miRNAs can negatively influence the expression of their downstream genes, thereby affecting the development of human cancers. The pathogenesis of acute lymphoblastic leukemia (ALL) is complex and more biomarkers and functional molecules need to be found. We attempted to reveal the specific mechanisms and functions of miRNA-181b-5p in ALL and investigated the effects of the miRNA-181b-5p/SSX2IP axis on ALL. Materials and Methods: Bioinformatics analyses were initially performed to screen out differentially expressed miRNAs in ALL and determine the research subject. qRT-PCR and western blotting were applied to evaluate the expression levels of target genes. Cell function experiments and mouse experiments were conducted to analyze the roles of the target genes in ALL. Results: miRNA-181b-5p was highly and differentially expressed in ALL and may target SSX2IP. The upregulation of miRNA-181b-5p and downregulation of SSX2IP were observed in ALL cells. miRNA-181b-5p could control multiple pathological processes of ALL, including cell proliferation, the cell cycle, and apoptosis, and miRNA-181b-5p could also facilitate tumor growth in vivo. Conclusion: miRNA-181b-5p promoted the malignant progression of ALL by downregulating SSX2IP. The miRNA-181b-5p/SSX2IP axis may be a promising target for intervention against the malignant behaviors of ALL.
Collapse
|
3
|
Liu X, Chen B, Chen J, Sun S. A novel tp53-associated nomogram to predict the overall survival in patients with pancreatic cancer. BMC Cancer 2021; 21:335. [PMID: 33789615 PMCID: PMC8011162 DOI: 10.1186/s12885-021-08066-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2020] [Accepted: 03/15/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Gene mutations play critical roles in tumorigenesis and cancer development. Our study aimed to screen survival-related mutations and explore a novel gene signature to predict the overall survival in pancreatic cancer. METHODS Somatic mutation data from three cohorts were used to identify the common survival-related gene mutation with Kaplan-Meier curves. RNA-sequencing data were used to explore the signature for survival prediction. First, Weighted Gene Co-expression Network Analysis was conducted to identify candidate genes. Then, the ICGC-PACA-CA cohort was applied as the training set and the TCGA-PAAD cohort was used as the external validation set. A TP53-associated signature calculating the risk score of every patient was developed with univariate Cox, least absolute shrinkage and selection operator, and stepwise regression analysis. Kaplan-Meier and receiver operating characteristic curves were plotted to verify the accuracy. The independence of the signature was confirmed by the multivariate Cox regression analysis. Finally, a prognostic nomogram including 359 patients was constructed based on the combined expression data and the risk scores. RESULTS TP53 mutation was screened to be the robust and survival-related mutation type, and was associated with immune cell infiltration. Two thousand, four hundred fifty-five genes included in the six modules generated in the WGCNA were screened as candidate survival related TP53-associated genes. A seven-gene signature was constructed: Risk score = (0.1254 × ERRFI1) - (0.1365 × IL6R) - (0.4400 × PPP1R10) - (0.3397 × PTOV1-AS2) + (0.1544 × SCEL) - (0.4412 × SSX2IP) - (0.2231 × TXNL4A). Area Under Curves of 1-, 3-, and 5-year ROC curves were 0.731, 0.808, and 0.873 in the training set and 0.703, 0.677, and 0.737 in the validation set. A prognostic nomogram including 359 patients was constructed and well-calibrated, with the Area Under Curves of 1-, 3-, and 5-year ROC curves as 0.713, 0.753, and 0.823. CONCLUSIONS The TP53-associated signature exhibited good prognostic efficacy in predicting the overall survival of PC patients.
Collapse
Affiliation(s)
- Xun Liu
- Department of Pancreas and Endocrine Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Bobo Chen
- Department of Pancreas and Endocrine Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Jiahui Chen
- Department of Pancreas and Endocrine Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China
| | - Shaolong Sun
- Department of Pancreas and Endocrine Surgery, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang, 110004, Liaoning, China.
| |
Collapse
|
4
|
Hori A, Toda T. Regulation of centriolar satellite integrity and its physiology. Cell Mol Life Sci 2016; 74:213-229. [PMID: 27484406 PMCID: PMC5219025 DOI: 10.1007/s00018-016-2315-x] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Revised: 07/14/2016] [Accepted: 07/21/2016] [Indexed: 01/01/2023]
Abstract
Centriolar satellites comprise cytoplasmic granules that are located around the centrosome. Their molecular identification was first reported more than a quarter of a century ago. These particles are not static in the cell but instead constantly move around the centrosome. Over the last decade, significant advances in their molecular compositions and biological functions have been achieved due to comprehensive proteomics and genomics, super-resolution microscopy analyses and elegant genetic manipulations. Centriolar satellites play pivotal roles in centrosome assembly and primary cilium formation through the delivery of centriolar/centrosomal components from the cytoplasm to the centrosome. Their importance is further underscored by the fact that mutations in genes encoding satellite components and regulators lead to various human disorders such as ciliopathies. Moreover, the most recent findings highlight dynamic structural remodelling in response to internal and external cues and unexpected positive feedback control that is exerted from the centrosome for centriolar satellite integrity.
Collapse
Affiliation(s)
- Akiko Hori
- Lincoln's Inn Fields Laboratory, The Francis Crick Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK.,Developmental Biomedical Science, Graduate School of Biological Sciences, Nara Institute of Science and Technology (NAIST), 8916-5 Takayama, Ikoma, Nara, 630-0192, Japan
| | - Takashi Toda
- Lincoln's Inn Fields Laboratory, The Francis Crick Institute, 44 Lincoln's Inn Fields, London, WC2A 3LY, UK. .,Department of Molecular Biotechnology, Hiroshima Research Center for Healthy Aging (HiHA), Graduate School of Advanced Sciences of Matter, Hiroshima University, 1-3-1 Kagamiyama, Higashi-Hiroshima, 739-8530, Japan.
| |
Collapse
|
5
|
Jones KB, Barrott JJ, Xie M, Haldar M, Jin H, Zhu JF, Monument MJ, Mosbruger TL, Langer EM, Randall RL, Wilson RK, Cairns BR, Ding L, Capecchi MR. The impact of chromosomal translocation locus and fusion oncogene coding sequence in synovial sarcomagenesis. Oncogene 2016; 35:5021-32. [PMID: 26947017 PMCID: PMC5014712 DOI: 10.1038/onc.2016.38] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 12/14/2015] [Accepted: 01/11/2016] [Indexed: 02/07/2023]
Abstract
Synovial sarcomas are aggressive soft-tissue malignancies that express chromosomal translocation-generated fusion genes, SS18-SSX1 or SS18-SSX2 in most cases. Here, we report a mouse sarcoma model expressing SS18-SSX1, complementing our prior model expressing SS18-SSX2. Exome sequencing identified no recurrent secondary mutations in tumors of either genotype. Most of the few mutations identified in single tumors were present in genes that were minimally or not expressed in any of the tumors. Chromosome 6, either entirely or around the fusion gene expression locus, demonstrated a copy number gain in a majority of tumors of both genotypes. Thus, by fusion oncogene coding sequence alone, SS18-SSX1 and SS18-SSX2 can each drive comparable synovial sarcomagenesis, independent from other genetic drivers. SS18-SSX1 and SS18-SSX2 tumor transcriptomes demonstrated very few consistent differences overall. In direct tumorigenesis comparisons, SS18-SSX2 was slightly more sarcomagenic than SS18-SSX1, but equivalent in its generation of biphasic histologic features. Meta-analysis of human synovial sarcoma patient series identified two tumor-gentoype-phenotype correlations that were not modeled by the mice, namely a scarcity of male hosts and biphasic histologic features among SS18-SSX2 tumors. Re-analysis of human SS18-SSX1 and SS18-SSX2 tumor transcriptomes demonstrated very few consistent differences, but highlighted increased native SSX2 expression in SS18-SSX1 tumors. This suggests that the translocated locus may drive genotype-phenotype differences more than the coding sequence of the fusion gene created. Two possible roles for native SSX2 in synovial sarcomagenesis are explored. Thus, even specific partial failures of mouse genetic modeling can be instructive to human tumor biology.
Collapse
Affiliation(s)
- K B Jones
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - J J Barrott
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - M Xie
- Department of Medicine, Washington University, St Louis, MO, USA
| | - M Haldar
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - H Jin
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - J-F Zhu
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - M J Monument
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - T L Mosbruger
- Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Department of Bioinformatics, University of Utah, Salt Lake City, UT, USA
| | - E M Langer
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| | - R L Randall
- Department of Orthopaedics, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - R K Wilson
- Department of Medicine, Washington University, St Louis, MO, USA.,McDonnell Genome Institute, Washington University, St Louis, MO, USA.,Department of Genetics, Washington University, St Louis, MO, USA.,Siteman Cancer Center, Washington University, St Louis, MO, USA
| | - B R Cairns
- Department of Oncological Sciences, University of Utah, Salt Lake City, UT, USA.,Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA.,Howard Hughes Medical Institute, University of Utah, Salt Lake City, UT, USA
| | - L Ding
- Department of Medicine, Washington University, St Louis, MO, USA.,McDonnell Genome Institute, Washington University, St Louis, MO, USA.,Department of Genetics, Washington University, St Louis, MO, USA.,Siteman Cancer Center, Washington University, St Louis, MO, USA
| | - M R Capecchi
- Department of Human Genetics, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
6
|
Peñagaricano F, Valente BD, Steibel JP, Bates RO, Ernst CW, Khatib H, Rosa GJM. Exploring causal networks underlying fat deposition and muscularity in pigs through the integration of phenotypic, genotypic and transcriptomic data. BMC SYSTEMS BIOLOGY 2015; 9:58. [PMID: 26376630 PMCID: PMC4574162 DOI: 10.1186/s12918-015-0207-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 09/04/2015] [Indexed: 12/23/2022]
Abstract
BACKGROUND Joint modeling and analysis of phenotypic, genotypic and transcriptomic data have the potential to uncover the genetic control of gene activity and phenotypic variation, as well as shed light on the manner and extent of connectedness among these variables. Current studies mainly report associations, i.e. undirected connections among variables without causal interpretation. Knowledge regarding causal relationships among genes and phenotypes can be used to predict the behavior of complex systems, as well as to optimize management practices and selection strategies. Here, we performed a multistep procedure for inferring causal networks underlying carcass fat deposition and muscularity in pigs using multi-omics data obtained from an F2 Duroc x Pietrain resource pig population. RESULTS We initially explored marginal associations between genotypes and phenotypic and expression traits through whole-genome scans, and then, in genomic regions with multiple significant hits, we assessed gene-phenotype network reconstruction using causal structural learning algorithms. One genomic region on SSC6 showed significant associations with three relevant phenotypes, off-midline10th-rib backfat thickness, loin muscle weight, and average intramuscular fat percentage, and also with the expression of seven genes, including ZNF24, SSX2IP, and AKR7A2. The inferred network indicated that the genotype affects the three phenotypes mainly through the expression of several genes. Among the phenotypes, fat deposition traits negatively affected loin muscle weight. CONCLUSIONS Our findings shed light on the antagonist relationship between carcass fat deposition and lean meat content in pigs. In addition, the procedure described in this study has the potential to unravel gene-phenotype networks underlying complex phenotypes.
Collapse
Affiliation(s)
- Francisco Peñagaricano
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Present Address: Department of Animal Sciences, and University of Florida Genetics Institute, University of Florida, Gainesville, FL, 326111, USA.
| | - Bruno D Valente
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Dairy Science, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Juan P Steibel
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA.
| | - Ronald O Bates
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA.
| | - Catherine W Ernst
- Department of Animal Science, Michigan State University, East Lansing, MI, 48824, USA.
| | - Hasan Khatib
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| | - Guilherme J M Rosa
- Department of Animal Sciences, University of Wisconsin-Madison, Madison, WI, 53706, USA.
- Biostatistics and Medical Informatics, University of Wisconsin-Madison, Madison, WI, 53706, USA.
| |
Collapse
|
7
|
Piwowar M, Jurkowski W. ONION: Functional Approach for Integration of Lipidomics and Transcriptomics Data. PLoS One 2015; 10:e0128854. [PMID: 26053255 PMCID: PMC4459700 DOI: 10.1371/journal.pone.0128854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 05/03/2015] [Indexed: 12/19/2022] Open
Abstract
To date, the massive quantity of data generated by high-throughput techniques has not yet met bioinformatics treatment required to make full use of it. This is partially due to a mismatch in experimental and analytical study design but primarily due to a lack of adequate analytical approaches. When integrating multiple data types e.g. transcriptomics and metabolomics, multidimensional statistical methods are currently the techniques of choice. Typical statistical approaches, such as canonical correlation analysis (CCA), that are applied to find associations between metabolites and genes are failing due to small numbers of observations (e.g. conditions, diet etc.) in comparison to data size (number of genes, metabolites). Modifications designed to cope with this issue are not ideal due to the need to add simulated data resulting in a lack of p-value computation or by pruning of variables hence losing potentially valid information. Instead, our approach makes use of verified or putative molecular interactions or functional association to guide analysis. The workflow includes dividing of data sets to reach the expected data structure, statistical analysis within groups and interpretation of results. By applying pathway and network analysis, data obtained by various platforms are grouped with moderate stringency to avoid functional bias. As a consequence CCA and other multivariate models can be applied to calculate robust statistics and provide easy to interpret associations between metabolites and genes to leverage understanding of metabolic response. Effective integration of lipidomics and transcriptomics is demonstrated on publically available murine nutrigenomics data sets. We are able to demonstrate that our approach improves detection of genes related to lipid metabolism, in comparison to applying statistics alone. This is measured by increased percentage of explained variance (95% vs. 75–80%) and by identifying new metabolite-gene associations related to lipid metabolism.
Collapse
Affiliation(s)
- Monika Piwowar
- Department of Bioinformatics and Telemedicine, Jagiellonian University, Kopernika 7E, 31–062 Kraków, Poland
| | - Wiktor Jurkowski
- The Genome Analysis Centre, Norwich Research Park, Norwich NR4 7UH, United Kingdom
- * E-mail:
| |
Collapse
|
8
|
Hori A, Peddie CJ, Collinson LM, Toda T. Centriolar satellite- and hMsd1/SSX2IP-dependent microtubule anchoring is critical for centriole assembly. Mol Biol Cell 2015; 26:2005-19. [PMID: 25833712 PMCID: PMC4472012 DOI: 10.1091/mbc.e14-11-1561] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/27/2015] [Indexed: 01/05/2023] Open
Abstract
Human Msd1/SSX2IP is a component of centriolar satellites and essential for microtubule anchoring to the centrosome. Anchoring defects lead to abnormal accumulation of centriolar components at centriolar satellites, which interferes with centriole assembly. Loss of Msd1/SSX2IP produces supernumerary centriole precursors specifically in tumor cells. Centriolar satellites are numerous electron-dense granules dispersed around the centrosome. Mutations in their components are linked to various human diseases, but their molecular roles remain elusive. In particular, the significance of spatial communication between centriolar satellites and the centrosome is unknown. hMsd1/SSX2IP localizes to both the centrosome and centriolar satellites and is required for tethering microtubules to the centrosome. Here we show that hMsd1/SSX2IP-mediated microtubule anchoring is essential for proper centriole assembly and duplication. On hMsd1/SSX2IP knockdown, the centriolar satellites become stuck at the microtubule minus end near the centrosome. Intriguingly, these satellites contain many proteins that normally localize to the centrosome. Of importance, microtubule structures, albeit not being anchored properly, are still required for the emergence of abnormal satellites, as complete microtubule depolymerization results in the disappearance of these aggregates from the vicinity of the centrosome. We highlighted, using superresolution and electron microscopy, that under these conditions, centriole structures are faulty. Remarkably, these cells are insensitive to Plk4 overproduction–induced ectopic centriole formation, yet they accelerate centrosome reduplication upon hydroxyurea arrest. Finally, the appearance of satellite aggregates is cancer cell specific. Together our findings provide novel insights into the mechanism of centriole assembly and microtubule anchoring.
Collapse
Affiliation(s)
- Akiko Hori
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Christopher J Peddie
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Lucy M Collinson
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| | - Takashi Toda
- The Francis Crick Institute, Lincoln's Inn Fields Laboratory, London WC2A 3LY, United Kingdom
| |
Collapse
|
9
|
Ikuno K, Kajii TS, Oka A, Inoko H, Ishikawa H, Iida J. Microsatellite genome-wide association study for mandibular prognathism. Am J Orthod Dentofacial Orthop 2014; 145:757-62. [PMID: 24880846 DOI: 10.1016/j.ajodo.2014.01.022] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2013] [Revised: 01/01/2014] [Accepted: 01/01/2014] [Indexed: 11/29/2022]
Abstract
INTRODUCTION Attempts have been made to identify susceptibility genes of mandibular prognathism by genome-wide linkage studies, but the results of susceptibility loci are inconsistent. There has been no genome-wide association study of mandibular prognathism. Our objective was to perform a genome-wide association study using 23,465 microsatellite markers to detect mandibular prognathism susceptibility regions. METHODS The study was based on the pooled DNA method, including 2 steps of screening on the whole genome and subsequent individual genotyping, with 240 experimental subjects and 360 control subjects from the Japanese population. RESULTS Two suggestive associations on chromosomes 1q32.2 (D1S1358i: P = 4.22 × 10(-4)) and 1p22.3 (D1S0411i: P = 6.66 × 10(-4)) were shown, and PLXNA2 and SSX2IP were suggested to be candidate genes; 1p22.3 flanked the region indicated by previous linkage analysis. CONCLUSIONS The results of the genome-wide association study showed that 2 loci (1q32.2 and 1p22.3) are likely to be susceptibility regions of mandibular prognathism: 1p32.2 is a novel locus, and identification of 1p22.3 supports the results of previous linkage analysis.
Collapse
Affiliation(s)
- Keiichiro Ikuno
- Postgraduate student, Division of Oral Functional Science, Department of Orthodontics, Graduate School of Dental Medicine, Hokkaido University, Hokkaido, Japan
| | - Takashi S Kajii
- Associate professor, Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan.
| | - Akira Oka
- Lecturer, Institute of Medical Science, Tokai University, Kanagawa, Japan
| | - Hidetoshi Inoko
- Professor, Division of Basic Medical Science and Molecular Medicine, Department of Molecular Life Sciences, School of Medicine, Tokai University, Kanagawa, Japan
| | - Hiroyuki Ishikawa
- Professor, Section of Orthodontics, Department of Oral Growth and Development, Fukuoka Dental College, Fukuoka, Japan
| | - Junichiro Iida
- Professor, Division of Oral Functional Science, Department of Orthodontics, Graduate School of Dental Medicine, Hokkaido University, Hokkaido, Japan
| |
Collapse
|
10
|
Li P, Lin Y, Zhang Y, Zhu Z, Huo K. SSX2IP promotes metastasis and chemotherapeutic resistance of hepatocellular carcinoma. J Transl Med 2013; 11:52. [PMID: 23452395 PMCID: PMC3599991 DOI: 10.1186/1479-5876-11-52] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2012] [Accepted: 12/14/2012] [Indexed: 12/31/2022] Open
Abstract
Background Synovial sarcoma, X breakpoint 2 interacting protein (SSX2IP), which has been identified as an acute myeloid leukemia associated antigen, is a potential target for leukemia immunotherapy. In rodents, its homologous gene, ADIP, plays an important role in the regulation of cell adhesion and migration, underlying its potential role in promoting metastasis of other cancers. Methods To investigate the correlation between the expression level of SSX2IP and the clinicopathologic factors of hepatocellular carcinoma (HCC), 53 cases were studied by qPCR and statisted. To directly testing SSX2IP’s contribution to HCC in animal models, 45 nude mice were enrolled in peritoneal spreading and liver metastasis models. For the migration and invasion assays, cell culture experiments were performed using QCMTM 24-Well Colorimetric Migration Assay Kit and Cell Invasion Assay Kit (Millipore). Moreover we examined the influence of SSX2IP overexpression on the chemosensitivity of hepatocellular carcinoma cells to two most common chemotherapy drugs (5-Fu and CDDP) using Cell counting kit-8 (CCK-8). The chemotherapeutic drugs sensitivity was evaluated by IC50 parameter. Results Statistical analysis of clinical cases revealed that the SSX2IP high expression group had inclinations towards larger tumor size, more tumor thrombus and shorter survival period, implying a strong correlation between the expression level of SSX2IP and HCC tumorigenesis. Consistently in abdominal cavity metastasis and liver metastasis models of immune-deficient mice, SSX2IP was able to promote the metastasis of hepatoma cells. At the cytological level, SSX2IP stimulates the wound healing, metastasis and invasion of hepatoma cells, and reduces the sensitivity of hepatoma cells to 5-Fu and CDDP. Conclusions Our results showed that SSX2IP promotes the development and metastasis of hepatocellular carcinoma and contributes to the drug resistance of hepatoma cells, suggesting that SSX2IP is expected to become a new diagnostic and prognostic marker and a new target of the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Pu Li
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, 220 Handan Rd, Shanghai 200433, People's Republic of China
| | | | | | | | | |
Collapse
|
11
|
The SSX family of cancer-testis antigens as target proteins for tumor therapy. Clin Dev Immunol 2010; 2010:150591. [PMID: 20981248 PMCID: PMC2963798 DOI: 10.1155/2010/150591] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2010] [Accepted: 09/01/2010] [Indexed: 01/01/2023]
Abstract
Cancer-testis antigens (CTAs) represent an expanding class of tumor-associated proteins defined on the basis of their tissue-restricted expression to testis or ovary germline cells and frequent ectopic expression in tumor tissue. The expression of CTA in MHC class I-deficient germline cells makes these proteins particularly attractive as immunotherapeutic targets because they serve as essentially tumor-specific antigens for MHC class I-restricted CD8+ T cells. Moreover, because CTAs are expressed in many types of cancer, any therapeutic developed to target these antigens might have efficacy for multiple cancer types. Of particular interest among CTAs is the synovial sarcoma X chromosome breakpoint (SSX) family of proteins, which includes ten highly homologous family members. Expression of SSX proteins in tumor tissues has been associated with advanced stages of disease and worse patient prognosis. Additionally, both humoral and cell-mediated immune responses to SSX proteins have been demonstrated in patients with tumors of varying histological origin, which indicates that natural immune responses can be spontaneously generated to these antigens in cancer patients. The current review will describe the history and identification of this family of proteins, as well as what is known of their function, expression in normal and malignant tissues, and immunogenicity.
Collapse
|
12
|
Voisin V, Legault P, Ospina DPS, Ben-David Y, Rassart E. Gene profiling of the erythro- and megakaryoblastic leukaemias induced by the Graffi murine retrovirus. BMC Med Genomics 2010; 3:2. [PMID: 20102610 PMCID: PMC2843641 DOI: 10.1186/1755-8794-3-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2009] [Accepted: 01/26/2010] [Indexed: 12/02/2022] Open
Abstract
Background Acute erythro- and megakaryoblastic leukaemias are associated with very poor prognoses and the mechanism of blastic transformation is insufficiently elucidated. The murine Graffi leukaemia retrovirus induces erythro- and megakaryoblastic leukaemias when inoculated into NFS mice and represents a good model to study these leukaemias. Methods To expand our understanding of genes specific to these leukaemias, we compared gene expression profiles, measured by microarray and RT-PCR, of all leukaemia types induced by this virus. Results The transcriptome level changes, present between the different leukaemias, led to the identification of specific cancerous signatures. We reported numerous genes that may be potential oncogenes, may have a function related to erythropoiesis or megakaryopoiesis or have a poorly elucidated physiological role. The expression pattern of these genes has been further tested by RT-PCR in different samples, in a Friend erythroleukaemic model and in human leukaemic cell lines. We also screened the megakaryoblastic leukaemias for viral integrations and identified genes targeted by these integrations and potentially implicated in the onset of the disease. Conclusions Taken as a whole, the data obtained from this global gene profiling experiment have provided a detailed characterization of Graffi virus induced erythro- and megakaryoblastic leukaemias with many genes reported specific to the transcriptome of these leukaemias for the first time.
Collapse
Affiliation(s)
- Veronique Voisin
- Laboratoire de Biologie Moléculaire, Département des Sciences Biologiques, Centre BioMed, Université du Québec à Montréal, Case Postale 8888 Succursale Centre-ville, Montréal, QC, Canada
| | | | | | | | | |
Collapse
|
13
|
Holterhus PM, Bebermeier JH, Werner R, Demeter J, Richter-Unruh A, Cario G, Appari M, Siebert R, Riepe F, Brooks JD, Hiort O. Disorders of sex development expose transcriptional autonomy of genetic sex and androgen-programmed hormonal sex in human blood leukocytes. BMC Genomics 2009; 10:292. [PMID: 19570224 PMCID: PMC2713997 DOI: 10.1186/1471-2164-10-292] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2009] [Accepted: 07/01/2009] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Gender appears to be determined by independent programs controlled by the sex-chromosomes and by androgen-dependent programming during embryonic development. To enable experimental dissection of these components in the human, we performed genome-wide profiling of the transcriptomes of peripheral blood mononuclear cells (PBMC) in patients with rare defined "disorders of sex development" (DSD, e.g., 46, XY-females due to defective androgen biosynthesis) compared to normal 46, XY-males and 46, XX-females. RESULTS A discrete set of transcripts was directly correlated with XY or XX genotypes in all individuals independent of male or female phenotype of the external genitalia. However, a significantly larger gene set in the PBMC only reflected the degree of external genital masculinization independent of the sex chromosomes and independent of concurrent post-natal sex steroid hormone levels. Consequently, the architecture of the transcriptional PBMC-"sexes" was either male, female or even "intersex" with a discordant alignment of the DSD individuals' genetic and hormonal sex signatures. CONCLUSION A significant fraction of gene expression differences between males and females in the human appears to have its roots in early embryogenesis and is not only caused by sex chromosomes but also by long-term sex-specific hormonal programming due to presence or absence of androgen during the time of external genital masculinization. Genetic sex and the androgen milieu during embryonic development might therefore independently modulate functional traits, phenotype and diseases associated with male or female gender as well as with DSD conditions.
Collapse
|
14
|
Samuel MS, Lundgren-May T, Ernst M. Identification of putative targets of DNA (cytosine-5) methylation-mediated transcriptional silencing using a novel conditionally active form of DNA methyltransferase 3a. Growth Factors 2007; 25:426-36. [PMID: 18365873 DOI: 10.1080/08977190801931081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
Aberrant DNA methylation of gene promoters is a recurrent finding associated with diseases such as cancer and inflammation, and is thought to contribute to disease through its role in transcriptional repression. Indeed, recent evidence suggests that DNA (cytosine-5) methyltransferases (DNMTs) may mediate the activity of factors promoting cell growth. Here, we utilise a novel experimental system for the conditional and reversible activation of a de novo DNMT by constructing a steroid-hormone analogue activated version, Dnmt3a-mERtrade mark. Following treatment with the oestrogen analogue 4-hydroxy tamoxifen of murine embryonic stem cells expressing this protein, we have identified by microarray analysis, several potential targets of Dnmt3a mediated transcriptional repression including the cancer associated genes Ssx2ip, Hmga1 and Wrnip. These results were validated using quantitative reverse transcriptase PCR and we confirm the biological significance of these in vitro observations by demonstrating a reduction in mRNA transcripts of the same genes within the intestinal epithelium of cancer-prone transgenic knock-in mutant mice over-expressing Dnmt3a throughout the intestinal epithelium.
Collapse
Affiliation(s)
- Michael S Samuel
- Ludwig Institute for Cancer Research, P. O. Royal Melbourne Hospital, Parkville, Vic., Australia
| | | | | |
Collapse
|