1
|
Cheng Q, Han X, Xie H, Liao YL, Wang F, Cui XY, Jiang , Zhang CW. PAXIP1 is regulated by NRF1 and is a prognosis‑related biomarker in hepatocellular carcinoma. Biomed Rep 2025; 22:38. [PMID: 39781045 PMCID: PMC11704871 DOI: 10.3892/br.2024.1916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is characterized by a poor prognosis globally. PAX-interacting protein 1 (PAXIP1) serves a key role in the development of numerous human cancer types. Nevertheless, its specific involvement in HCC remains poorly understood. Public repository systems (Integrative Molecular Database of HCC, Gene Expression Omnibus, The Cancer Genome Atlas, University of Alabama at Birmingham Cancer Data Analysis Portal, Tumor Immune Estimation Resource and Human Protein Atlas) were utilized to explore PAXIP1 expression in HCC and evaluate the prognostic value of PAXIP1 in patients with HCC. PAXIP1 expression was investigated, and a notable relationship between PAXIP1 expression and various cancer types was found through analysis of The Cancer Genome Atlas data. More specifically, patients with HCC and lower PAXIP1 levels had improved survival rates. Furthermore, using LinkedOmics, the co-expression network of PAXIP1 in HCC was determined. Colocalization analysis of PAXIP1 using chromatin immunoprecipitation-sequencing data suggested that PAXIP1 might act as a cofactor for MYB proto-oncogene like 2 or FOXO1 in HCC. In addition, by predicting and analyzing the potential transcription factors related to PAXIP1, nuclear respiratory factor 1 was identified as a factor upstream of PAXIP1 in HCC. Notably, PAXIP1 expression exhibited a positive association with the infiltration of CD4+ and CD8+ T cells, macrophages, neutrophils and myeloid dendritic cells. Furthermore, PAXIP1 expression was associated with a range of immune markers such as programmed cell death protein 1, programmed death-ligand 1 and cytotoxic T-lymphocyte associated protein 4 in HCC. The findings of the present study highlighted the prognostic relevance of PAXIP1 and its function in modulating immune cell recruitment in HCC.
Collapse
Affiliation(s)
- Qian Cheng
- Department of Pathogen Biology, Microbiology Division, Key Laboratory of Pathogen of Jiangsu Province Nanjing Medical University, Nanjing, Jiangsu 211166, P.R. China
| | - Xiao Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Hao Xie
- School of Life Science and Technology, Southeast University, Nanjing, Jiangsu 210018, P.R. China
| | - Yan-Lin Liao
- MEDx (Suzhou) Translation Medicine Co., Ltd., Suzhou, Jiangsu 215000, P.R. China
| | - Fei Wang
- Wuxi Mental Health Center/Wuxi Central Rehabilitation Hospital, Wuxi School of Medicine, Jiangnan University, Wuxi, Jiangsu 214122, P.R. China
| | - Xiao-Ying Cui
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu 210029, P.R. China
| | - Chao Jiang
- Department of Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| | - Cheng-Wan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu 223300, P.R. China
| |
Collapse
|
2
|
Rodrigues DB, Moreira HR, Jarnalo M, Horta R, Marques AP, Reis RL, Pirraco RP. Generation of 3D melanoma models using an assembloid-based approach. Acta Biomater 2024; 178:93-110. [PMID: 38382833 DOI: 10.1016/j.actbio.2024.02.023] [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: 07/25/2023] [Revised: 01/31/2024] [Accepted: 02/13/2024] [Indexed: 02/23/2024]
Abstract
While 3D tumor models have greatly evolved over the past years, there is still a strong requirement for more biosimilar models which are capable of recapitulating cellular crosstalk within the tumor microenvironment while equally displaying representative levels of tumor aggressiveness and invasion. Herein, we disclose an assembloid melanoma model based on the fusion of individual stromal multicellular spheroids (MCSs). In contrast to more traditional tumor models, we show that it is possible to develop self-organizing, heterotypic melanoma models where tumor cells present stem-cell like features like up-regulated pluripotency master regulators SOX2, POU5F1 and NANOG. Additionally, these assembloids display high levels of invasiveness while embedded in 3D matrices as evidenced by stromal cell promotion of melanoma cell invasion via metalloproteinase production. Furthermore, sensitivity to anticancer drug doxorubicin was demonstrated for the melanoma assembloid model. These findings suggest that melanoma assembloids may play a significant role in the field of 3D cancer models as they more closely mimic the tumor microenvironment when compared to more traditional MCSs, opening the doors to a better understanding of the role of tumor microenvironment in supporting tumor progression. STATEMENT OF SIGNIFICANCE: The development of complex 3D tumor models that better recapitulate the tumor microenvironment is crucial for both an improved comprehension of intercellular crosstalk and for more efficient drug screening. We have herein developed a self-organizing heterotypic assembloid-based melanoma model capable of closely mimicking the tumor microenvironment. Key features recapitulated were the preservation of cancer cell stemness, sensitivity to anti-cancer agents and tumor cell invasion promoted by stromal cells. The approach of pre-establishing distinct stromal domains for subsequent combination into more complex tumor constructs provides a route for developing superior tumor models with a higher degree of similarity to native cancer tissues.
Collapse
Affiliation(s)
- Daniel B Rodrigues
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Helena R Moreira
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Mariana Jarnalo
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, Porto, Portugal; Faculty of Medicine - University of Porto, Portugal
| | - Ricardo Horta
- Department of Plastic and Reconstructive Surgery, and Burn Unity, Centro Hospitalar de São João, Porto, Portugal; Faculty of Medicine - University of Porto, Portugal
| | - Alexandra P Marques
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Rui L Reis
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal
| | - Rogério P Pirraco
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's - PT Government Associate Laboratory, Braga, Guimarães 4805-017, Portugal.
| |
Collapse
|
3
|
Zhang X, Li Y, Chen Y. Development of a Comprehensive Gene Signature Linking Hypoxia, Glycolysis, Lactylation, and Metabolomic Insights in Gastric Cancer through the Integration of Bulk and Single-Cell RNA-Seq Data. Biomedicines 2023; 11:2948. [PMID: 38001949 PMCID: PMC10669360 DOI: 10.3390/biomedicines11112948] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/17/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
BACKGROUND Hypoxia and anaerobic glycolysis are cancer hallmarks and sources of the metabolite lactate. Intriguingly, lactate-induced protein lactylation is considered a novel epigenetic mechanism that predisposes cells toward a malignant state. However, the significance of comprehensive hypoxia-glycolysis-lactylation-related genes (HGLRGs) in cancer is unclear. We aimed to construct a model centered around HGLRGs for predicting survival, metabolic features, drug responsiveness, and immune response in gastric cancer. METHODS The integration of bulk and single-cell RNA-Seq data was achieved using data obtained from the TCGA and GEO databases to analyze HGLRG expression patterns. A HGLRG risk-score model was developed based on univariate Cox regression and a LASSO-Cox regression model and subsequently validated. Additionally, the relationships between the identified HGLRG signature and multiple metabolites, drug sensitivity and various cell clusters were explored. RESULTS Thirteen genes were identified as constituting the HGLRG signature. Using this signature, we established predictive models, including HGLRG risk scores and nomogram and Cox regression models. The stratification of patients into high- and low-risk groups based on HGLRG risk scores showed a better prognosis in the latter. The high-risk group displayed increased sensitivity to cytotoxic drugs and targeted inhibitors. The expression of the HGLRG BGN displayed a strong correlation with amino acids and lipid metabolites. Notably, a significant difference in immune infiltration, such as that of M1 macrophages and CD8 T cells, was correlated with the HGLRG signature. The abundant DUSP1 within the mesenchymal components was highlighted by single-cell transcriptomics. CONCLUSION The innovative HGLRG signature demonstrates efficacy in predicting survival and providing a practical clinical model for gastric cancer. The HGLRG signature reflects the internal metabolism, drug responsiveness, and immune microenvironment components of gastric cancer and is expected to boost patients' response to targeted therapy and immunotherapy.
Collapse
Affiliation(s)
- Xiangqian Zhang
- NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yun Li
- NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Yongheng Chen
- NHC Key Laboratory of Cancer Proteomics & State Local Joint Engineering Laboratroy for Anticancer Drugs, Department of Oncology, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
4
|
Peña-Guerrero J, Fernández-Rubio C, García-Sosa AT, Nguewa PA. BRCT Domains: Structure, Functions, and Implications in Disease-New Therapeutic Targets for Innovative Drug Discovery against Infections. Pharmaceutics 2023; 15:1839. [PMID: 37514027 PMCID: PMC10386641 DOI: 10.3390/pharmaceutics15071839] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/12/2023] [Accepted: 06/22/2023] [Indexed: 07/30/2023] Open
Abstract
The search for new therapeutic targets and their implications in drug development remains an emerging scientific topic. BRCT-bearing proteins are found in Archaea, Bacteria, Eukarya, and viruses. They are traditionally involved in DNA repair, recombination, and cell cycle control. To carry out these functions, BRCT domains are able to interact with DNA and proteins. Moreover, such domains are also implicated in several pathogenic processes and malignancies including breast, ovarian, and lung cancer. Although these domains exhibit moderately conserved folding, their sequences show very low conservation. Interestingly, sequence variations among species are considered positive traits in the search for suitable therapeutic targets, since non-specific drug interactions might be reduced. These main characteristics of BRCT, as well as its critical implications in key biological processes in the cell, have prompted the study of these domains as therapeutic targets. This review explores the possible roles of BRCT domains as therapeutic targets for drug discovery. We describe their common structural features and relevant interactions and pathways, as well as their implications in pathologic processes. Drugs commonly used to target these domains are also presented. Finally, based on their structures, we describe new drug design possibilities using modern and innovative techniques.
Collapse
Affiliation(s)
- José Peña-Guerrero
- ISTUN Institute of Tropical Health, Department of Microbiology and Parasitology, University of Navarra, IdiSNA (Navarra Institute for Health Research), E-31008 Pamplona, Navarra, Spain
| | - Celia Fernández-Rubio
- ISTUN Institute of Tropical Health, Department of Microbiology and Parasitology, University of Navarra, IdiSNA (Navarra Institute for Health Research), E-31008 Pamplona, Navarra, Spain
| | - Alfonso T García-Sosa
- Chair of Molecular Technology, Institute of Chemistry, University of Tartu, Ravila 14a, 50411 Tartu, Estonia
| | - Paul A Nguewa
- ISTUN Institute of Tropical Health, Department of Microbiology and Parasitology, University of Navarra, IdiSNA (Navarra Institute for Health Research), E-31008 Pamplona, Navarra, Spain
| |
Collapse
|
5
|
DNA Double-Strand Break-Related Competitive Endogenous RNA Network of Noncoding RNA in Bovine Cumulus Cells. Genes (Basel) 2023; 14:genes14020290. [PMID: 36833217 PMCID: PMC9956238 DOI: 10.3390/genes14020290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
(1) Background: DNA double strand breaks (DSBs) are the most serious form of DNA damage that affects oocyte maturation and the physiological state of follicles and ovaries. Non-coding RNAs (ncRNAs) play a crucial role in DNA damage and repair. This study aims to analyze and establish the network of ncRNAs when DSB occurs and provide new ideas for next research on the mechanism of cumulus DSB. (2) Methods: Bovine cumulus cells (CCs) were treated with bleomycin (BLM) to construct a DSB model. We detected the changes of the cell cycle, cell viability, and apoptosis to determine the effect of DSBs on cell biology, and further evaluated the relationship between the transcriptome and competitive endogenous RNA (ceRNA) network and DSBs. (3) Results: BLM increased γH2AX positivity in CCs, disrupted the G1/S phase, and decreased cell viability. Totals of 848 mRNAs, 75 long noncoding RNAs (lncRNAs), 68 circular RNAs (circRNAs), and 71 microRNAs (miRNAs) in 78 groups of lncRNA-miRNA-mRNA regulatory networks, 275 groups of circRNA-miRNA-mRNA regulatory networks, and five groups of lncRNA/circRNA-miRNA-mRNA co-expression regulatory networks were related to DSBs. Most differentially expressed ncRNAs were annotated to cell cycle, p53, PI3K-AKT, and WNT signaling pathways. (4) Conclusions: The ceRNA network helps to understand the effects of DNA DSBs activation and remission on the biological function of CCs.
Collapse
|
6
|
Liu Y, Han J, Kong T, Xiao N, Mei Q, Liu J. DriverMP enables improved identification of cancer driver genes. Gigascience 2022; 12:giad106. [PMID: 38091511 PMCID: PMC10716827 DOI: 10.1093/gigascience/giad106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 10/30/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
BACKGROUND Cancer is widely regarded as a complex disease primarily driven by genetic mutations. A critical concern and significant obstacle lies in discerning driver genes amid an extensive array of passenger genes. FINDINGS We present a new method termed DriverMP for effectively prioritizing altered genes on a cancer-type level by considering mutated gene pairs. It is designed to first apply nonsilent somatic mutation data, protein‒protein interaction network data, and differential gene expression data to prioritize mutated gene pairs, and then individual mutated genes are prioritized based on prioritized mutated gene pairs. Application of this method in 10 cancer datasets from The Cancer Genome Atlas demonstrated its great improvements over all the compared state-of-the-art methods in identifying known driver genes. Then, a comprehensive analysis demonstrated the reliability of the novel driver genes that are strongly supported by clinical experiments, disease enrichment, or biological pathway analysis. CONCLUSIONS The new method, DriverMP, which is able to identify driver genes by effectively integrating the advantages of multiple kinds of cancer data, is available at https://github.com/LiuYangyangSDU/DriverMP. In addition, we have developed a novel driver gene database for 10 cancer types and an online service that can be freely accessed without registration for users. The DriverMP method, the database of novel drivers, and the user-friendly online server are expected to contribute to new diagnostic and therapeutic opportunities for cancers.
Collapse
Affiliation(s)
- Yangyang Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Jiyun Han
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Tongxin Kong
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Nannan Xiao
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| | - Qinglin Mei
- MOE Key Laboratory of Bioinformatics, BNRIST Bioinformatics Division, Department of Automation, Tsinghua University, Beijing 100084, China
| | - Juntao Liu
- School of Mathematics and Statistics, Shandong University (Weihai), Weihai 264209, China
| |
Collapse
|
7
|
Wang W, Ou Z, Peng J, Wang N, Zhou Y. Bioinformatics-based analysis of potential candidates chromatin regulators for immune infiltration in osteoarthritis. BMC Musculoskelet Disord 2022; 23:1123. [PMID: 36550476 PMCID: PMC9783407 DOI: 10.1186/s12891-022-06098-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Through the bioinformatics analysis to screen out the potential chromatin regulators (CRs) under the immune infiltration of osteoarthritis (OA), thus providing some theoretical support for future studies of epigenetic mechanisms under OA immune infiltration. METHODS By integrating CRs and the OA gene expression matrix, we performed weighted gene co-expression network analysis (WGCNA), differential analysis, and further screened Hub genes by protein-protein interaction (PPI) analysis. Using the OA gene expression matrix, immune infiltration extraction and quantification were performed to analyze the correlations and differences between immune infiltrating cells and their functions. By virtue of these Hub genes, Hub gene association analysis was completed and their upstream miRNAs were predicted by the FunRich software. Moreover, a risk model was established to analyze the risk probability of associated CRs in OA, and the confidence of the results was validated by the validation dataset. RESULTS This research acquired a total of 32 overlapping genes, and 10 Hub genes were further identified. The strongest positive correlation between dendritic cells and mast cells and the strongest negative correlation between parainflammation and Type I IFN reponse. In the OA group DCs, iDCs, macrophages, MCs, APC co-inhibition, and CCR were significantly increased, whereas B cells, NK cells, Th2 cells, TIL, and T cell co-stimulation were significantly decreased. The risk model results revealed that BRD1 might be an independent risk factor for OA, and the validation dataset results are consistent with it. 60 upstream miRNAs of OA-related CRs were predicted by the FunRich software. CONCLUSION This study identified certain potential CRs and miRNAs that could regulate OA immunity, thus providing certain theoretical supports for future epigenetic mechanism studies on the immune infiltration of OA.
Collapse
Affiliation(s)
- Weiwei Wang
- Guilin Hospital of Traditional Chinese Medicine, Guilin, 541002 Guangxi China
| | - Zhixue Ou
- Guilin Hospital of Traditional Chinese Medicine, Guilin, 541002 Guangxi China
| | - Jianlan Peng
- grid.256609.e0000 0001 2254 5798Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530001 Guangxi China
| | - Ning Wang
- grid.511973.8The First Affiliated Hospital of Guangxi University of Traditional Chinese Medicine, Nanning, 530001 Guangxi China
| | - Yi Zhou
- grid.256609.e0000 0001 2254 5798Ruikang Hospital Affiliated to Guangxi University of Traditional Chinese Medicine, Nanning, 530001 Guangxi China
| |
Collapse
|
8
|
Möller S, Saul N, Projahn E, Barrantes I, Gézsi A, Walter M, Antal P, Fuellen G. Gene co-expression analyses of health(span) across multiple species. NAR Genom Bioinform 2022; 4:lqac083. [PMID: 36458022 PMCID: PMC9706456 DOI: 10.1093/nargab/lqac083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Revised: 08/20/2022] [Accepted: 10/31/2022] [Indexed: 12/03/2022] Open
Abstract
Health(span)-related gene clusters/modules were recently identified based on knowledge about the cross-species genetic basis of health, to interpret transcriptomic datasets describing health-related interventions. However, the cross-species comparison of health-related observations reveals a lot of heterogeneity, not least due to widely varying health(span) definitions and study designs, posing a challenge for the exploration of conserved healthspan modules and, specifically, their transfer across species. To improve the identification and exploration of conserved/transferable healthspan modules, here we apply an established workflow based on gene co-expression network analyses employing GEO/ArrayExpress data for human and animal models, and perform a comprehensive meta-study of the resulting modules related to health(span), yielding a small set of literature backed health(span) candidate genes. For each experiment, WGCNA (weighted gene correlation network analysis) was used to infer modules of genes which correlate in their expression with a 'health phenotype score' and to determine the most-connected (hub) genes (and their interactions) for each such module. After mapping these hub genes to their human orthologs, 12 health(span) genes were identified in at least two species (ACTN3, ANK1, MRPL18, MYL1, PAXIP1, PPP1CA, SCN3B, SDCBP, SKIV2L, TUBG1, TYROBP, WIPF1), for which enrichment analysis by g:profiler found an association with actin filament-based movement and associated organelles, as well as muscular structures. We conclude that a meta-study of hub genes from co-expression network analyses for the complex phenotype health(span), across multiple species, can yield molecular-mechanistic insights and can direct experimentalists to further investigate the contribution of individual genes and their interactions to health(span).
Collapse
Affiliation(s)
- Steffen Möller
- To whom correspondence should be addressed. Tel: +49 381 494 7361; Fax: +49 381 494 7203;
| | - Nadine Saul
- Humboldt-University of Berlin, Institute of Biology, Berlin, Germany
| | - Elias Projahn
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - Israel Barrantes
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| | - András Gézsi
- Budapest University of Technology and Economics, Department of Measurement and Information Systems, Budapest, Hungary
| | - Michael Walter
- Rostock University Medical Center, Institute for Clinical Chemistry and Laboratory Medicine, Rostock, Germany
| | - Péter Antal
- Budapest University of Technology and Economics, Department of Measurement and Information Systems, Budapest, Hungary
| | - Georg Fuellen
- Rostock University Medical Center, Institute for Biostatistics and Informatics in Medicine and Ageing Research, Rostock, Germany
| |
Collapse
|
9
|
Zhang F, Wei M, Chen H, Ji L, Nie Y, Kang J. The genomic stability regulator PTIP is required for proper chromosome segregation in mitosis. Cell Div 2022; 17:5. [PMID: 36153541 PMCID: PMC9509598 DOI: 10.1186/s13008-022-00081-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 09/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background The Pax transcription activation domain-interacting protein (PTIP) is a nuclear protein that is an essential component of H3K4 methylation for gene activation in vascular, kidney, B cell, and adipocyte development. Furthermore, it plays a key role in genomic stability in higher eukaryotic cells. It binds to 53BP1 and antagonizes inappropriate homologous recombination for a proper DNA damage response. Interestingly, an early study reported mitotic defects after PTIP inactivation, but it is not clear whether PTIP directly facilitates mitotic processes. Results Here, we showed that PTIP is essential for the mitotic integrity of HeLa cells. PTIP inactivation increases cell death during mitotic exit, which appears to result from direct mitotic defects. PTIP inactivation did not affect the G2M DNA damage checkpoint during interphase upon etoposide treatment. However, in mitosis, PTIP inactivation results in prolonged mitotic time, inefficient chromosome alignment, and increased cell death. Furthermore, PTIP localizes to the mitotic centrosome via BRCT domains at the C-terminus. Conclusion This study reveals a novel function of PTIP in maintaining the genomic stability of higher eukaryotes during mitosis. Therefore, its deregulation, which occurs in various tumors, may destabilize the genome by introducing an abnormal DNA damage response, as well as erroneous chromosome segregation. Supplementary Information The online version contains supplementary material available at 10.1186/s13008-022-00081-4.
Collapse
|
10
|
Zhang QX, Yang Y, Yang H, Guo Q, Guo JL, Liu HS, Zhang J, Li D. The roles of risk model based on the 3-XRCC genes in lung adenocarcinoma progression. Transl Cancer Res 2022; 10:4413-4431. [PMID: 35116299 PMCID: PMC8798971 DOI: 10.21037/tcr-21-1431] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/26/2021] [Indexed: 02/05/2023]
Abstract
Background The abnormal expression of deoxyribonucleic acid (DNA) repair genes might be the cause of tumor development and resistance of malignant cells to chemotherapeutic drugs. A risk model based on the X-ray repair of cross-complementary (XRCC) genes was constructed to improve the diagnosis and treatment of lung adenocarcinoma (LUAD) patients. Methods The expression levels, diagnostic values, and prognostic values of XRCC genes were identified, and the roles and regulatory mechanisms of the risk model based on the XRCC4/5/6 in LUAD progression was explored via The Cancer Genome Atlas (TCGA) and Oncomine databases. Results XRCC1/2/3/4/5/6, XRCC7 (PRKDC), and XRCC9 (FANCG) were overexpressed, and had diagnostic value for LUAD. The XRCC genes were involved in DNA repair, and participated in the regulation of non-homologous end-joining, homologous recombination, etc. The overall survival (OS), tumor (T) stage, and survival status of patients were significantly different between the Cluster1 and Cluster2 groups. XRCC4/5/6 were independent risk factors affecting the prognosis of LUAD patients. The risk score was related to the prognosis, sex, clinical stage, T, lymph node (N), and metastasis (M) stage, as well as the survival status of LUAD patients. The clinical stage and risk score were independent risk factors for poor prognosis in LUAD patients. The risk model was involved in RNA degradation, cell cycle, basal transcription factors, DNA replication etc. The risk scores were significantly correlated with the expression levels of TGFBR1, CD160, TNFSF4, TNFRSF14, IL6R, CXCL16, TNFRSF25, TAPBP, CCL16, and CCL14. Conclusions The risk model based on the XRCC4/5/6 genes could predict the progression of LUAD patients.
Collapse
Affiliation(s)
- Qun-Xian Zhang
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Ye Yang
- Department of Psychiatry, Traditional Chinese Medicine Hospital of Shiyan, Shiyan, China
| | - Heng Yang
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,Postgraduate Training Basement of Jinzhou Medical University, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Qiang Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Jia-Long Guo
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,Postgraduate Training Basement of Jinzhou Medical University, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Hua-Song Liu
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Jun Zhang
- Department of Cardiothoracic Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Dan Li
- Department of Oncology, Huanggang Central Hospital, Huanggang, China
| |
Collapse
|
11
|
Zhang L, Wei XT, Niu JJ, Lin ZX, Xu Q, Ni JJ, Zhang WL, Han BX, Yan SS, Feng GJ, Zhang H, Yang XL, Zhang ZJ, Hai R, Ren HG, Zhang F, Pei YF. Joint Genome-Wide Association Analyses Identified 49 Novel Loci For Age at Natural Menopause. J Clin Endocrinol Metab 2021; 106:2574-2591. [PMID: 34050765 DOI: 10.1210/clinem/dgab377] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Indexed: 01/07/2023]
Abstract
BACKGROUND Age at natural menopause (ANM) is an important index for women's health. Either early or late ANM is associated with a series of adverse outcomes later in life. Despite being an inheritable trait, its genetic determinant has not yet been fully understood. METHODS Aiming to better characterize the genetic architecture of ANM, we conducted genome-wide association study (GWAS) meta-analyses in European-specific as well as trans-ancestry samples by using GWAS summary statistics from the following 3 large studies: the Reproductive Genetics Consortium (ReproGen; N = 69 626), the UK Biobank cohort (UKBB; N = 111 593) and the BioBank Japan Project (BBJ; N = 43 861), followed by a series of bioinformatical assessments and functional annotations. RESULTS By integrating the summary statistics from the 3 GWAS of up to 225 200 participants, this largest meta-analysis identified 49 novel loci and 3 secondary signals that were associated with ANM at the genome-wide significance level (P < 5 × 10-8). No population specificity or heterogeneity was observed at most of the associated loci. Functional annotations prioritized 90 candidate genes at the newly identified loci. Among the 26 traits that were genetically correlated with ANM, hormone replacement therapy (HRT) exerted a causal relationship, implying a causal pattern by which HRT was determined by ANM. CONCLUSION Our findings improved our understanding of the etiology of female menopause, as well as shed light on potential new therapies for abnormal menopause.
Collapse
Affiliation(s)
- Lei Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Xin-Tong Wei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Jun-Jie Niu
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
| | - Zi-Xuan Lin
- Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Qian Xu
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Jing-Jing Ni
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Wan-Lin Zhang
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Bai-Xue Han
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Shan-Shan Yan
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Gui-Juan Feng
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Hong Zhang
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College, Soochow University, Suzhou, China
| | - Xiao-Lin Yang
- Center for Genetic Epidemiology and Genomics, School of Public Health, Medical College, Soochow University, Suzhou, China
- Department of Gastroenterology, Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, Yangzhou, China
| | - Zi-Jia Zhang
- Health Commission of Inner Mongolia Autonomous Region, Inner Mongolia Autonomous Region, Hohhot, China
| | - Rong Hai
- Health Commission of Inner Mongolia Autonomous Region, Inner Mongolia Autonomous Region, Hohhot, China
| | - Hai-Gang Ren
- Jiangsu Key laboratory of Translational Research and Therapy for Neuropsychiatric disorders & Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Feng Zhang
- Key Laboratory of Trace Elements and Endemic Diseases of National Health and Family Planning Commission, School of Public Health, Health Science Center, Xi'an Jiaotong University, Xi'an, China
| | - Yu-Fang Pei
- Department of Epidemiology and Health Statistics, School of Public Health, Medical College, the first affiliated hospital of Soochow University, Suzhou, China
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, School of Public Health, Medical College, Soochow University, Suzhou, China
| |
Collapse
|
12
|
McInerney-Leo AM, Chew HY, Inglis PL, Leo PJ, Joseph SR, Cooper CL, Okano S, Hassall T, Anderson L, Bowman RV, Gattas M, Harris JE, Marshall MS, Shaw JG, Wheeler L, Yang IA, Brown MA, Fong KM, Simpson F, Duncan EL. Germline ERBB3 mutation in familial non-small cell lung carcinoma: Expanding ErbB's role in oncogenesis. Hum Mol Genet 2021; 30:2393-2401. [PMID: 34274969 PMCID: PMC8643496 DOI: 10.1093/hmg/ddab172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 06/12/2021] [Accepted: 06/21/2021] [Indexed: 11/22/2022] Open
Abstract
Lung cancer is the commonest cause of cancer deaths worldwide. Although strongly associated with smoking, predisposition to lung cancer is also heritable, with multiple common risk variants identified. Rarely, dominantly inherited non-small-cell lung cancer (NSCLC) has been reported due to somatic mutations in EGFR/ErbB1 and ERBB2. Germline exome sequencing was performed in a multi-generation family with autosomal dominant NSCLC, including an affected child. Tumour samples were also sequenced. Full-length wild-type (wtErbB3) and mutant ERBB3 (mutErbB3) constructs were transfected into HeLa cells. Protein expression, stability, and subcellular localization were assessed, and cellular proliferation, pAkt/Akt and pERK levels determined. A novel germline variant in ERBB3 (c.1946 T > G: p.Iso649Arg), coding for receptor tyrosine-protein kinase erbB-3 (ErbB3), was identified, with appropriate segregation. There was no loss-of-heterozygosity in tumour samples. Both wtErbB3 and mutErbB3 were stably expressed. MutErbB3-transfected cells demonstrated an increased ratio of the 80 kDa form (which enhances proliferation) compared with the full-length (180 kDa) form. MutErbB3 and wtErbB3 had similar punctate cytoplasmic localization pre- and post-epidermal growth factor stimulation; however, epidermal growth factor receptor (EGFR) levels decreased faster post-stimulation in mutErbB3-transfected cells, suggesting more rapid processing of the mutErbB3/EGFR heterodimer. Cellular proliferation was increased in mutErbB3-transfected cells compared with wtErbB3 transfection. MutErbB3-transfected cells also showed decreased pAkt/tAkt ratios and increased pERK/tERK 30 min post-stimulation compared with wtErbB3 transfection, demonstrating altered signalling pathway activation. Cumulatively, these results support this mutation as tumorogenic. This is the first reported family with a germline ERBB3 mutation causing heritable NSCLC, furthering understanding of the ErbB family pathway in oncogenesis.
Collapse
Affiliation(s)
- Aideen M McInerney-Leo
- The Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102
| | - Hui Yi Chew
- The Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102
| | - Po-Ling Inglis
- Medical Oncology, Royal Brisbane and Women's Hospital, Herston, QLD, 4029
| | - Paul J Leo
- Australian Translational Genomics Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102
| | - Shannon R Joseph
- The Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102
| | - Caroline L Cooper
- Department of Anatomical Pathology, Pathology Queensland, Princess Alexandra Hospital, Brisbane.,UQTRC, Faculty of Medicine, The University of Queensland, 288 Herston Road, Herston, QLD, 4006
| | - Satomi Okano
- The Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102
| | - Tim Hassall
- Queensland Children's Hospital, South Brisbane, QLD, 4101
| | - Lisa Anderson
- Medical Oncology, Royal Brisbane and Women's Hospital, Herston, QLD, 4029
| | - Rayleen V Bowman
- UQTRC, Faculty of Medicine, The University of Queensland, 288 Herston Road, Herston, QLD, 4006.,Department of Thoracic Medicine, The Prince Charles Hospital, Rode Road, Chermside, QLD, 4032
| | - Michael Gattas
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Herston, QLD, 4029
| | - Jessica E Harris
- Australian Translational Genomics Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102
| | - Mhairi S Marshall
- Australian Translational Genomics Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102
| | - Janet G Shaw
- UQTRC, Faculty of Medicine, The University of Queensland, 288 Herston Road, Herston, QLD, 4006.,Department of Thoracic Medicine, The Prince Charles Hospital, Rode Road, Chermside, QLD, 4032
| | - Lawrie Wheeler
- Australian Translational Genomics Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102
| | - Ian A Yang
- UQTRC, Faculty of Medicine, The University of Queensland, 288 Herston Road, Herston, QLD, 4006.,Department of Thoracic Medicine, The Prince Charles Hospital, Rode Road, Chermside, QLD, 4032
| | - Matthew A Brown
- Australian Translational Genomics Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102.,Guy's and St Thomas' NHS Foundation Trust, London, United Kingdom.,King's College London NIHR Biomedical Research Centre, King's College London, United Kingdom
| | - Kwun M Fong
- UQTRC, Faculty of Medicine, The University of Queensland, 288 Herston Road, Herston, QLD, 4006.,Department of Thoracic Medicine, The Prince Charles Hospital, Rode Road, Chermside, QLD, 4032
| | - Fiona Simpson
- The Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD, 4102
| | - Emma L Duncan
- Australian Translational Genomics Centre, Institute of Health and Biomedical Innovation, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, 37 Kent St, Woolloongabba, QLD, 4102.,UQTRC, Faculty of Medicine, The University of Queensland, 288 Herston Road, Herston, QLD, 4006.,Department of Twin Research and Genetic Epidemiology, Faculty of Life Sciences and Medicine, King's College London, United Kingdom
| |
Collapse
|
13
|
Han X, Zhu Y, Shen L, Zhou Y, Pang L, Zhou W, Gu H, Han K, Yang Y, Jiang C, Xie J, Zhang C, Ding L. PTIP Inhibits Cell Invasion in Esophageal Squamous Cell Carcinoma via Modulation of EphA2 Expression. Front Oncol 2021; 11:629916. [PMID: 33833989 PMCID: PMC8021923 DOI: 10.3389/fonc.2021.629916] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Accepted: 03/05/2021] [Indexed: 12/30/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is a highly aggressive malignancy and treatment failure is largely due to metastasis and invasion. Aberrant tumor cell adhesion is often associated with tumor progression and metastasis. However, the exact details of cell adhesion in ESCC progression have yet to be determined. In our study, the clinical relevance of Pax2 transactivation domain-interacting protein (PTIP/PAXIP1) was analyzed by immunohistochemistry of ESCC tissues. We found that low expression of PTIP was associated with lymph node metastasis in ESCC, and loss-of-function approaches showed that depletion of PTIP promoted ESCC cell migration and invasion both in vitro and in vivo. Analysis integrating RNA-seq and ChIP-seq data revealed that PTIP directly regulated ephrin type-A receptor 2 (EphA2) expression in ESCC cells. Moreover, PTIP inhibited EphA2 expression by competing with Fosl2, which attenuated the invasion ability of ESCC cells. These results collectively suggest that PTIP regulates ESCC invasion through modulation of EphA2 expression and hence presents a potential therapeutic target for its treatment.
Collapse
Affiliation(s)
- Xiao Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yaning Zhu
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Li Shen
- Department of Hematology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yu Zhou
- Department of Medical Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Liqun Pang
- Department of General Surgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Wubi Zhou
- Department of Pathology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Hao Gu
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Kairong Han
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Yijun Yang
- Department of Obstetrics and Gynecology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Chao Jiang
- Department of Medical Oncology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Jun Xie
- Department of Clinical Laboratory, Xuyi People's Hospital, Huai'an, China
| | - Chengwan Zhang
- Department of Central Laboratory, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| | - Lianshu Ding
- Department of Neurosurgery, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huai'an, China
| |
Collapse
|
14
|
Pan-cancer analysis of genomic properties and clinical outcome associated with tumor tertiary lymphoid structure. Sci Rep 2020; 10:21530. [PMID: 33299035 PMCID: PMC7725838 DOI: 10.1038/s41598-020-78560-3] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/17/2020] [Indexed: 12/13/2022] Open
Abstract
How the genomic landscape of a tumor shapes the formation of tertiary lymphoid structure (TLS) and how might TLS alter the clinical outcome or response to immunotherapy had not been systematically explored. Utilizing the genomic and transcriptome data of solid tumors on TCGA, we quantified TLS based on a previous identified 12-chemokine signature and evaluated its correlation with mutation/neoantigen burden, functional mutation of oncogenes and the presence of viral infection. Clinical data was integrated to decide the prognostic significance of TLS for different cancers after surgical treatment. Publicly available data (clinical and transcriptome data) of immunotherapy clinical trials involving melanoma and lung cancer were also collected to evaluate TLS’s association with therapeutic outcome. Mutation burden and predicted neoantigen counts were positively correlated with TLS scoring in multiple cancer types. Mutation in tumor suppressor genes (KEAP1, PBRM1) and genes involved in extrinsic apoptosis (CASP8), antigen-presentation (HLA-A, HLA-B), immune regulation (SMAD4) or DNA repair (BRCA1, BRCA2, TP53BP1) correlated with TLS alteration in multiple tumor types, indicating the interaction between mutation landscape and TLS formation. Epstein-Barr virus (EBV) infection in gastric cancer and human papillomavirus (HPV) infection in Head and Neck squamous cell carcinoma were associated with increased TLS scoring. High TLS scoring predicted favorable prognosis in certain cancer after surgical treatment and improved response to immunotherapy in lung cancer and melanoma. Our findings unraveled the genomic properties associated with TLS formation in different solid tumors and highlighted the prognostic and predictive significance of TLS in surgical treatment and immunotherapy.
Collapse
|
15
|
Gzil A, Jaworski D, Antosik P, Zarębska I, Durślewicz J, Dominiak J, Kasperska A, Neska-Długosz I, Grzanka D, Szylberg Ł. The impact of TP53BP1 and MLH1 on metastatic capability in cases of locally advanced prostate cancer and their usefulness in clinical practice. Urol Oncol 2020; 38:600.e17-600.e26. [PMID: 32280038 DOI: 10.1016/j.urolonc.2020.02.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Revised: 01/30/2020] [Accepted: 02/09/2020] [Indexed: 11/29/2022]
Abstract
BACKGROUND Lymph node (LN) metastases increase the risk of death from prostate cancer (CaP). The dysfunction of factors responsible for DNA injury detection may promote the evolution of localized primary tumors into the metastatic form. METHODS In this study, 52 cases of CaP were analyzed. The cases were divided into groups of CaP without metastases (N0), with metastases to the LNs (N+), and metastatic LN tissue. Immunohistochemical examinations were performed with antibodies against MDC1, TP53BP1, MLH1, MSH2, MSH6, and PMS2. RESULTS Statistical analysis showed lower nuclear expression of TP53BP1 in N+ cases than in N0 cases (P = 0.026). Nuclear TP53BP1 expression was lower in LN cases than in N+ cases (P = 0.019). Statistical analysis showed lower nuclear expression of MLH1 in N+ cases than in to N0 cases (P = 0.003). CONCLUSION Decreased expression of both MLH1 and TP53B1 were demonstrated in N+ cases of CaP. This observation could help to determine the risk of nodal metastasis, and to select appropriate treatment modalities for patients with locally advanced CaP.
Collapse
Affiliation(s)
- Arkadiusz Gzil
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland.
| | - Damian Jaworski
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Paulina Antosik
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Izabela Zarębska
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Justyna Durślewicz
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Joanna Dominiak
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Anna Kasperska
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Izabela Neska-Długosz
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Dariusz Grzanka
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland
| | - Łukasz Szylberg
- Department of Clinical Pathomorphology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Poland; Department of Pathomorphology, Military Clinical Hospital, Bydgoszcz, Poland; Department of Tumor Pathology and Pathomorphology, Oncology Center, Prof., Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
| |
Collapse
|
16
|
53BP1 Accumulation in Circulating Tumor Cells Identifies Chemotherapy-Responsive Metastatic Breast Cancer Patients. Cancers (Basel) 2020; 12:cancers12040930. [PMID: 32283863 PMCID: PMC7226269 DOI: 10.3390/cancers12040930] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/03/2020] [Accepted: 04/07/2020] [Indexed: 12/17/2022] Open
Abstract
Evidence suggests that the DNA end-binding protein p53-binding protein 1 (53BP1) is down-regulated in subsets of breast cancer. Circulating tumor cells (CTCs) provide accessible “biopsy material” to track cell traits and functions and their alterations during treatment. Here, we prospectively monitored the 53BP1 status in CTCs from 67 metastatic breast cancer (MBC) patients with HER2- CTCs and known hormone receptor (HR) status of the primary tumor and/or metastases before, during, and at the end of chemotherapeutic treatment with Eribulin. Nuclear 53BP1 staining and genomic integrity were evaluated by immunocytochemical and whole-genome-amplification-based polymerase chain reaction (PCR) analysis, respectively. Comparative analysis of CTCs from patients with triple-negative and HR+ tumors revealed elevated 53BP1 levels in CTCs from patients with HR+ metastases, particularly following chemotherapeutic treatment. Differences in nuclear 53BP1 signals did not correlate with genomic integrity in CTCs at baseline or with nuclear γH2AX signals in MBC cell lines, indicating that 53BP1 detected features beyond DNA damage. Kaplan–Meier analysis revealed an increasing association between nuclear 53BP1-positivity and progression-free survival (PFS) during chemotherapy until the final visit. Our data suggest that 53BP1 detection in CTCs could be a useful marker to capture dynamic changes of chemotherapeutic responsiveness in triple-negative and HR+ MBC.
Collapse
|
17
|
Xu H, Zhao G, Zhang Y, Jiang H, Wang W, Zhao D, Yu H, Qi L. Long non-coding RNA PAXIP1-AS1 facilitates cell invasion and angiogenesis of glioma by recruiting transcription factor ETS1 to upregulate KIF14 expression. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2019; 38:486. [PMID: 31823805 PMCID: PMC6902534 DOI: 10.1186/s13046-019-1474-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 11/06/2019] [Indexed: 12/29/2022]
Abstract
Background Gliomas are common life-threatening cancers, mainly due to their aggressive nature and frequent invasiveness and long non-coding RNAs (lncRNAs) are emerging as promising molecular targets. Therefore, we explored the regulatory mechanisms underlying the putative involvement of the lncRNA PAX-interacting protein 1- antisense RNA1/ETS proto-oncogene 1/kinesin family member 14 (PAXIP1-AS1/ETS1/KIF14) axis in glioma cell invasion and angiogenesis. Methods Firstly, we identified differentially expressed lncRNA PAXIP1-AS1 as associated with glioma based on bioinformatic data. Then, validation experiments were conducted to confirm a high expression level of lncRNA PAXIP1-AS1 in glioma tissues and cells, accompanied by upregulated KIF14. We further examined the binding between lncRNA PAXIP1-AS1, KIF14 promoter activity, and transcription factor ETS1. Next, overexpression vectors and shRNAs were delivered to alter the expression of lncRNA PAXIP1-AS1, KIF14, and ETS1 to analyze their effects on glioma progression in vivo and in vitro. Results LncRNA PAXIP1-AS1 was mainly distributed in the nucleus of glioma cells. LncRNA PAXIP1-AS1 could upregulate the KIF14 promoter activity by recruiting transcription factor ETS1. Overexpression of lncRNA PAXIP1-AS1 enhanced migration, invasion, and angiogenesis of human umbilical vein endothelial cells in glioma by recruiting the transcription factor ETS1 to upregulate the expression of KIF14, which was further confirmed by accelerated tumor growth in nude mice. Conclusions The key findings of this study highlighted the potential of the lncRNA PAXIP1-AS1/ETS1/KIF14 axis as a therapeutic target for glioma treatment, due to its role in controlling the migration and invasion of glioma cells and its angiogenesis.
Collapse
Affiliation(s)
- Haiyang Xu
- Department of Oncological Neurosurgery, First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China
| | - Guifang Zhao
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, B24 Yinquan South Road, Qingyuan, 511518, Guang dong Province, People's Republic of China.,Department of Pathophysiology, Jilin Medical University, No. 5, Jilin Street, Jilin, 132013, Jilin Province, People's Republic of China
| | - Yu Zhang
- Department of Neurovascular, First Hospital of Jilin University, Changchun, 130021, People's Republic of China
| | - Hong Jiang
- Department of Ophthalmology, China-Japan Union Hospital of Jilin University, Changchun, 130033, People's Republic of China
| | - Weiyao Wang
- Department of Pathophysiology, Jilin Medical University, No. 5, Jilin Street, Jilin, 132013, Jilin Province, People's Republic of China
| | - Donghai Zhao
- Department of Pathophysiology, Jilin Medical University, No. 5, Jilin Street, Jilin, 132013, Jilin Province, People's Republic of China
| | - Hongquan Yu
- Department of Oncological Neurosurgery, First Hospital of Jilin University, No. 71, Xinmin Street, Changchun, 130021, Jilin Province, People's Republic of China.
| | - Ling Qi
- Qingyuan People's Hospital, The Sixth Affiliated Hospital of Guangzhou Medical University, B24 Yinquan South Road, Qingyuan, 511518, Guang dong Province, People's Republic of China. .,Department of Pathophysiology, Jilin Medical University, No. 5, Jilin Street, Jilin, 132013, Jilin Province, People's Republic of China.
| |
Collapse
|
18
|
Zhang X, Hu F, Liu L, Xu B. Effect of silencing of mediator of DNA damage checkpoint protein 1 on the growth of oral squamous cell carcinoma in vitro and in vivo. Eur J Oral Sci 2019; 127:494-499. [PMID: 31786813 DOI: 10.1111/eos.12662] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2019] [Indexed: 12/24/2022]
Abstract
Mediator of DNA damage checkpoint protein 1 (MDC1) is involved in DNA damage repair and has been linked to tumor invasion, metastasis, and prognosis. This study investigated the effects of MDC1 in oral squamous cell carcinoma (OSCC) in vitro and in vivo. RNA interference-mediated knockdown of MDC1 was performed in two OSCC cell lines (Tca-8113 and KB). Real-time PCR and western blotting were performed to determine expression of mRNA and protein, respectively, of MDC1. Cell viability was assessed using the MTT assay. Colony-formation assays were performed by staining with 0.5% crystal violet. Cell migration and invasion were detected by Transwell assays. The role of MDC1 in OSCC was examined in vivo via injection of Tca-8113 cells transfected with MDC1 small interfering (si)RNA or negative-control siRNA into a mouse xenograft model of OSCC. Our results showed that MDC1 knockdown decreased cell proliferation. Inhibition of MDC1 decreased colony formation of Tca-8113 and KB cells by 62% and 68%, respectively, and MDC1 knockdown reduced the number of migratory and invasive cells compared with the control group. Moreover, the xenograft mouse model of MDC1 knockdown showed reduced tumor growth. Our study suggests that MDC1 plays a role in tumorigenesis and might be a potential target for the treatment of patients with OSCC.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Fengling Hu
- Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Liuhui Liu
- Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| | - Bin Xu
- Department of Stomatology, The Fifth People's Hospital of Shanghai, Fudan University, Shanghai, China
| |
Collapse
|
19
|
53BP1: A key player of DNA damage response with critical functions in cancer. DNA Repair (Amst) 2019; 73:110-119. [DOI: 10.1016/j.dnarep.2018.11.008] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 11/18/2018] [Accepted: 11/19/2018] [Indexed: 02/06/2023]
|