1
|
Laub V, Nan E, Elias L, Donaldson IJ, Bentsen M, Rusling LA, Schupp J, Lun JH, Plate KH, Looso M, Langer JD, Günther S, Bobola N, Schulte D. Integrated multi-omics analysis of PBX1 in mouse adult neural stem- and progenitor cells identifies a transcriptional module that functionally links PBX1 to TCF3/4. Nucleic Acids Res 2024; 52:12262-12280. [PMID: 39377397 PMCID: PMC11551771 DOI: 10.1093/nar/gkae864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 08/22/2024] [Accepted: 09/23/2024] [Indexed: 10/09/2024] Open
Abstract
Developmental transcription factors act in networks, but how these networks achieve cell- and tissue specificity is still poorly understood. Here, we explored pre-B cell leukemia homeobox 1 (PBX1) in adult neurogenesis combining genomic, transcriptomic, and proteomic approaches. ChIP-seq analysis uncovered PBX1 binding to numerous genomic sites. Integration of PBX1 ChIP-seq with ATAC-seq data predicted interaction partners, which were subsequently validated by mass spectrometry. Whole transcriptome spatial RNA analysis revealed shared expression dynamics of Pbx1 and interacting factors. Among these were class I bHLH proteins TCF3 and TCF4. RNA-seq following Pbx1, Tcf3 or Tcf4 knockdown identified proliferation- and differentiation associated genes as shared targets, while sphere formation assays following knockdown argued for functional cooperativity of PBX1 and TCF3 in progenitor cell proliferation. Notably, while physiological PBX1-TCF interaction has not yet been described, chromosomal translocation resulting in genomic TCF3::PBX1 fusion characterizes a subtype of acute lymphoblastic leukemia. Introducing Pbx1 into Nalm6 cells, a pre-B cell line expressing TCF3 but lacking PBX1, upregulated the leukemogenic genes BLK and NOTCH3, arguing that functional PBX1-TCF cooperativity likely extends to hematopoiesis. Our study hence uncovers a transcriptional module orchestrating the balance between progenitor cell proliferation and differentiation in adult neurogenesis with potential implications for leukemia etiology.
Collapse
Affiliation(s)
- Vera Laub
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| | - Elisabeth Nan
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| | - Lena Elias
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| | - Ian J Donaldson
- University of Manchester, Faculty of Biology, Medicine and Health, Bioinformatics Core Facility, Manchester, M13 9PT, UK
| | - Mette Bentsen
- Max Planck Institute for Heart and Lung Research, Bioinformatics Core Unit (BCU), 61231 Bad Nauheim, Germany
| | - Leona A Rusling
- Max Planck Institute for Biophysics, Proteomics, and Max Planck Institute for Brain Research, 60438 Frankfurt am Main, Germany
| | - Jonathan Schupp
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
- Goethe University, Frankfurt Cancer Institute, 60528 Frankfurt am Main, Germany
| | - Jennifer H Lun
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
- Goethe University, Frankfurt Cancer Institute, 60528 Frankfurt am Main, Germany
| | - Karl H Plate
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
- Goethe University, Frankfurt Cancer Institute, 60528 Frankfurt am Main, Germany
| | - Mario Looso
- Max Planck Institute for Heart and Lung Research, Bioinformatics Core Unit (BCU), 61231 Bad Nauheim, Germany
| | - Julian D Langer
- Max Planck Institute for Biophysics, Proteomics, and Max Planck Institute for Brain Research, 60438 Frankfurt am Main, Germany
| | - Stefan Günther
- Max Planck Institute for Heart and Lung Research, Bioinformatics and Deep Sequencing Platform, 61231 Bad Nauheim, Germany
| | - Nicoletta Bobola
- University of Manchester, Faculty of Biology, Medicine and Health, Manchester, M13 9PT, UK
| | - Dorothea Schulte
- Goethe University, University Hospital Frankfurt, Neurological Institute (Edinger Institute), 60528 Frankfurt am Main, Germany
| |
Collapse
|
2
|
Estevam DD, Souza JM, Rey FSB, Martins CL, Stafuzza NB, Espigolan R, Millen DD, Arrigoni MDB. Identification of genomic regions and pathways associated with traits related to rumen acidosis in feedlot Nellore cattle. J Anim Breed Genet 2024; 141:491-506. [PMID: 38375946 DOI: 10.1111/jbg.12860] [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: 10/26/2022] [Revised: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 02/21/2024]
Abstract
There may be an increased risk of metabolic disorders, such as rumen acidosis, in cattle fed high-concentrate diets, particularly those from Bos taurus indicus genotypes, which have shown to be more sensitive to ruminal acidification. Therefore, this study aimed to estimate (co)variance components and identify genomic regions and pathways associated with ruminal acidosis in feedlot Nellore cattle fed high-concentrate diets. It was utilized a dataset containing a total of 642 Nellore bulls that were genotyped from seven feedlot nutrition studies. The GGP Indicus 35k panel was used with the single step genome-wide association study methodology in which the effects of the markers were obtained from the genomic values estimated by the GBLUP model. A bivariate model to estimate genetic correlations between the economically important traits and indicator traits for acidosis was used. The traits evaluated in this study that were nutritionally related to rumen acidosis included average daily gain (ADG), final body weight, time spent eating (TSE), time spent ruminating, rumenitis score (RUM), rumen absorptive surface area (ASA), rumen keratinized layer thickness (KER) and hot carcass weight (HCW). The identified candidate genes were mainly involved in the negative or non-regulation of the apoptotic process, salivary secretion, and transmembrane transport. The genetic correlation between HCW and ASA was low positive (0.27 ± 0.23), and between ADG and ASA was high moderate (0.58 ± 0.59). A positive genetic correlation between RUM and all performance traits was observed, and TSE correlated negatively with HCW (-0.33 ± 0.21), ASA (-0.75 ± 0.48), and KER (-0.40 ± 0.27). The genetic association between economically important traits and indicator traits for acidosis suggested that Nellore cattle may be more sensitive to acidosis in feedlot systems.
Collapse
Affiliation(s)
- Daniela D Estevam
- Department of Animal Production, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Johnny M Souza
- Department of Animal Science, College of Technology and Agricultural Sciences, São Paulo State University (UNESP), Dracena, São Paulo, Brazil
| | - Fernando S B Rey
- Department of Animal Science, School of Agricultural and Veterinary Studies, São Paulo State University (UNESP), Jaboticabal, São Paulo, Brazil
| | - Cyntia L Martins
- Department of Animal Production, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| | - Nedenia B Stafuzza
- Department of Animal Science, Animal Science Institute (IZ), São Paulo's Agency for Agribusiness Technology (APTA), Sertãozinho, São Paulo, Brazil
| | - Rafael Espigolan
- Department of Animal Science and Biological Sciences (DZCB), Federal University of Santa Maria (UFSM), Palmeira das Missões, Brazil
| | - Danilo D Millen
- Department of Animal Science, College of Technology and Agricultural Sciences, São Paulo State University (UNESP), Dracena, São Paulo, Brazil
| | - Mario D B Arrigoni
- Department of Animal Production, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu, São Paulo, Brazil
| |
Collapse
|
3
|
Fu J, Chen Y, Li J, Tan M, Lin R, Wang J, Wu G, Rao Y, Wu F, Gao Y, Bai M, Wang P, Wu F. Construction and analysis of a lysosome-dependent cell death score-based prediction model for non-small cell lung cancer. Discov Oncol 2024; 15:388. [PMID: 39212757 PMCID: PMC11364741 DOI: 10.1007/s12672-024-01233-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/06/2024] [Accepted: 08/13/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Non-small cell lung cancer (NSCLC) is the most common type of tumor globally and the leading cause of cancer-related deaths. Although treatment strategies such as immune checkpoint inhibitors and chemotherapy have advanced, the heterogeneity among NSCLC patients results in significant variability in treatment outcomes. Studies have shown that certain patients respond poorly to immune checkpoint inhibitors, indicating that treatment response is closely related to multiple factors. Therefore, it is necessary to develop predictive models to stratify patients based on gene expression and clinical characteristics, aiming for precision therapy. OBJECTIVE This study aims to construct a stratified prognostic model for NSCLC patients based on lysosome-dependent cell death (LDCD) scoring by integrating single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing data. By analyzing the immune-related characteristics of high-risk and low-risk groups, we further explored the impact of cell death patterns on lung cancer and identified potential therapeutic targets. METHODS This study obtained single-cell RNA sequencing data and gene expression data of NSCLC patients and normal lung tissues from the GEO and TCGA databases. We used R packages such as Seurat and CellChat for data preprocessing and analysis, and performed dimensionality reduction and visualization through Principal Component Analysis (PCA) and UMAP algorithms. LASSO regression analysis was used to construct the predictive model, followed by cross-validation and ROC curve analysis. The model's effectiveness was validated through survival analysis and immune microenvironment analysis. RESULTS The study showed a significant increase in the proportion of monocytes in NSCLC tissues, suggesting their important role in cancer progression. Cell communication analysis indicated that macrophages, smooth muscle cells, and myeloid cells exhibit strong intercellular communication during cancer progression. Using the constructed prognostic model based on 12 LDCD-related genes, we found significant differences in overall survival and immune microenvironment between the high-risk and low-risk groups.
Collapse
Affiliation(s)
- Jiangping Fu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China
| | - Yaohua Chen
- Department of General Respiratory, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jie Li
- Department of Clinical Research Center, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Ming Tan
- Department of Otolaryngology-Head and Neck Surgery, The Central Hospital of Jingmen, Jingmen, China
| | - Rui Lin
- Department of Oncology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Jiang Wang
- Department of Oncology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Guirong Wu
- Department of Oncology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Yao Rao
- Department of General Respiratory, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Fudao Wu
- Department of Oncology, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Youshu Gao
- Department of Ultrasound Imaging, Dazhou Central Hospital, Dazhou, Sichuan, China
| | - Maoshu Bai
- Department of Oncology, Dazhou Second People's Hospital, Dazhou Integrated Traditional Chinese Medicine and Western Medicine Hospital, Dazhou, Sichuan, China
| | - Pingfei Wang
- Department of General Respiratory, Dazhou Central Hospital, Dazhou, Sichuan, China.
| | - Fang Wu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi, China.
| |
Collapse
|
4
|
Magrath JW, Espinosa-Cotton M, Flinchum DA, Sampath SS, Cheung NK, Lee SB. Desmoplastic small round cell tumor: from genomics to targets, potential paths to future therapeutics. Front Cell Dev Biol 2024; 12:1442488. [PMID: 39139449 PMCID: PMC11319132 DOI: 10.3389/fcell.2024.1442488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 06/26/2024] [Indexed: 08/15/2024] Open
Abstract
Desmoplastic Small Round Cell Tumor (DSRCT) is a highly aggressive pediatric cancer caused by a reciprocal translocation between chromosomes 11 and 22, leading to the formation of the EWSR1::WT1 oncoprotein. DSRCT presents most commonly in the abdominal and pelvic peritoneum and remains refractory to current treatment regimens which include chemotherapy, radiotherapy, and surgery. As a rare cancer, sample and model availability have been a limiting factor to DSRCT research. However, the establishment of rare tumor banks and novel cell lines have recently propelled critical advances in the understanding of DSRCT biology and the identification of potentially promising targeted therapeutics. Here we review model and dataset availability, current understanding of the EWSR1::WT1 oncogenic mechanism, and promising preclinical therapeutics, some of which are now advancing to clinical trials. We discuss efforts to inhibit critical dependencies including NTRK3, EGFR, and CDK4/6 as well as novel immunotherapy strategies targeting surface markers highly expressed in DSRCT such as B7-H3 or neopeptides either derived from or driven by the fusion oncoprotein. Finally, we discuss the prospect of combination therapies and strategies for prioritizing clinical translation.
Collapse
Affiliation(s)
- Justin W. Magrath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Madelyn Espinosa-Cotton
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Dane A. Flinchum
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Shruthi Sanjitha Sampath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| | - Nai Kong Cheung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Sean B. Lee
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, LA, United States
| |
Collapse
|
5
|
Magrath JW, Flinchum DA, Hartono AB, Sampath SS, O'Grady TM, Baddoo M, Haoyang L, Xu X, Flemington EK, Lee SB. Transcriptomic analysis identifies B-lymphocyte kinase as a therapeutic target for desmoplastic small round cell tumor cancer stem cell-like cells. Oncogenesis 2024; 13:2. [PMID: 38177125 PMCID: PMC10767073 DOI: 10.1038/s41389-023-00504-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 11/28/2023] [Accepted: 12/07/2023] [Indexed: 01/06/2024] Open
Abstract
Desmoplastic small round cell tumor (DSRCT) is an aggressive pediatric cancer caused by the EWSR1-WT1 fusion oncoprotein. The tumor is refractory to treatment with a 5-year survival rate of only 15-25%, necessitating the development of novel therapeutics, especially those able to target chemoresistant subpopulations. Novel in vitro cancer stem cell-like (CSC-like) culture conditions increase the expression of stemness markers (SOX2, NANOG) and reduce DSRCT cell line susceptibility to chemotherapy while maintaining the ability of DSRCT cells to form xenografts. To gain insights into this chemoresistant model, RNA-seq was performed to elucidate transcriptional alterations between DSRCT cells grown in CSC-like spheres and normal 2-dimensional adherent state. Commonly upregulated and downregulated genes were identified and utilized in pathway analysis revealing upregulation of pathways related to chromatin assembly and disassembly and downregulation of pathways including cell junction assembly and extracellular matrix organization. Alterations in chromatin assembly suggest a role for epigenetics in the DSRCT CSC-like state, which was further investigated with ATAC-seq, identifying over 10,000 differentially accessible peaks, including 4444 sphere accessible peaks and 6,120 adherent accessible peaks. Accessible regions were associated with higher gene expression, including increased accessibility of the CSC marker SOX2 in CSC-like culture conditions. These analyses were further utilized to identify potential CSC therapeutic targets, leading to the identification of B-lymphocyte kinase (BLK) as a CSC-enriched, EWSR1-WT1-regulated, druggable target. BLK inhibition and knockdown reduced CSC-like properties, including abrogation of tumorsphere formation and stemness marker expression. Importantly, BLK knockdown reduced DSRCT CSC-like cell chemoresistance, making its inhibition a promising target for future combination therapy.
Collapse
Affiliation(s)
- Justin W Magrath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Dane A Flinchum
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Alifiani B Hartono
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
- Department of Molecular & Medical Pharmacology, University of California Los Angeles, 630 Charles E Young Dr. S., Los Angeles, CA, 90095, USA
| | - Shruthi Sanjitha Sampath
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Tina M O'Grady
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Melody Baddoo
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Liang Haoyang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Xiaojiang Xu
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Erik K Flemington
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA
| | - Sean B Lee
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, 1430 Tulane Ave, New Orleans, LA, USA.
| |
Collapse
|
6
|
Ralli S, Jones SJ, Leach S, Lynch HT, Brooks-Wilson AR. Gene and pathway based burden analyses in familial lymphoid cancer cases: Rare variants in immune pathway genes. PLoS One 2023; 18:e0287602. [PMID: 37379307 PMCID: PMC10306212 DOI: 10.1371/journal.pone.0287602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 06/08/2023] [Indexed: 06/30/2023] Open
Abstract
Genome-wide association studies have revealed common genetic variants with small effect sizes associated with diverse lymphoid cancers. Family studies have uncovered rare variants with high effect sizes. However, these variants explain only a portion of the heritability of these cancers. Some of the missing heritability may be attributable to rare variants with small effect sizes. We aim to identify rare germline variants associated with familial lymphoid cancers using exome sequencing. One case per family was selected from 39 lymphoid cancer families based on early onset of disease or rarity of subtype. Control data was from Non-Finnish Europeans in gnomAD exomes (N = 56,885) or ExAC (N = 33,370). Gene and pathway-based burden tests for rare variants were performed using TRAPD. Five putatively pathogenic germline variants were found in four genes: INTU, PEX7, EHHADH, and ASXL1. Pathway-based association tests identified the innate and adaptive immune systems, peroxisomal pathway and olfactory receptor pathway as associated with lymphoid cancers in familial cases. Our results suggest that rare inherited defects in the genes involved in immune system and peroxisomal pathway may predispose individuals to lymphoid cancers.
Collapse
Affiliation(s)
- Sneha Ralli
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| | - Samantha J. Jones
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Stephen Leach
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Henry T. Lynch
- Hereditary Cancer Center, Creighton University, Omaha, Nebraska, United States of America
| | - Angela R. Brooks-Wilson
- Department of Biomedical Physiology and Kinesiology, Simon Fraser University, Burnaby, British Columbia, Canada
- Canada’s Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, British Columbia, Canada
| |
Collapse
|
7
|
Xu Y, Xu J, Qiao R, Zhong H, Xia J, Zhong R. Loss of BLK expression as a potential predictor of poor prognosis and immune checkpoint blockade response in NSCLC and contribute to tumor progression. Transl Oncol 2023; 33:101671. [PMID: 37068401 PMCID: PMC10127141 DOI: 10.1016/j.tranon.2023.101671] [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: 01/06/2023] [Revised: 03/16/2023] [Accepted: 04/08/2023] [Indexed: 04/19/2023] Open
Abstract
BACKGROUND Immune checkpoint blockade (ICB) has been proved to have significant anti-tumor effect in the clinical treatment of non-small cell lung cancer (NSCLC). Therefore, biomarkers predicting ICB response can provide better treatment for patients with NSCLC. METHODS Differential expression genes (DEGs) were identified by ImmuCellAI database. Copy number alteration (CNA) was analyzed by cBioPortal. The predicted efficiency of 4 genes on cancer immunotherapy was assessed by ROC analysis. The survival value of BLK was analyzed by Kaplan-Meier plotter and Prognoscan analysis. Clinical significance of BLK IHC-TMA score in NSCLC was also explored. The CCK-8 assay, wound healing assay, western blot assay in vitro and subcutaneous xenograft experiments in vivo were used for investigating the functions of BLK. The RNA-sequencing were performed to screen BLK regulated genes and conducted for GO/KEGG enrichment analysis. The transcriptional regulatory factor of BLK promoter region was predicted by ChIP-seq analysis. RESULTS 39 common DEGs between ICB Response (R) group and No Response (NR) group with NSCLC were identified, in which the CNA frequency of BLK deletion (> 6%) was found. The predicted efficiency of BLK on immunotherapy was performed best in NSCLC (AUC>0.7). Low expression of BLK was related to NSCLC with significantly poor prognosis. BLK overexpression can inhibit growth of NSCLC via activating apoptosis pathway, inhibiting the G2M checkpoint and Glycolysis pathway. The enrichment analysis indicated that BLK regulated genes related to oncogenic potential in NSCLC. Besides, BLK expression was inhibited via H3K27me3 modification in A549 and H1299 cells. BLK mRNA level was negatively correlated with methylation and positively correlated with the tumor purity in NSCLC. CONCLUSION Our study provides strong evidence that low expression of BLK may serve as a biomarker for poor prognosis in NSCLC, while response to ICB therapy and contributes to NSCLC tumor progression.
Collapse
Affiliation(s)
- Yingqi Xu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 Huaihai West Road, Shanghai 200030, China.
| | - Jianlin Xu
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 Huaihai West Road, Shanghai 200030, China.
| | - Rong Qiao
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 Huaihai West Road, Shanghai 200030, China.
| | - Hua Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 Huaihai West Road, Shanghai 200030, China.
| | - Jinjing Xia
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 Huaihai West Road, Shanghai 200030, China.
| | - Runbo Zhong
- Department of Pulmonary Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 Huaihai West Road, Shanghai 200030, China.
| |
Collapse
|
8
|
Pasvenskaite A, Liutkeviciene R, Gedvilaite G, Vilkeviciute A, Liutkevicius V, Uloza V. The survival rate of laryngeal squamous cell carcinoma: impact of IL1RAP rs4624606, IL1RL1 rs1041973, IL-6 rs1800795, BLK rs13277113, and TIMP3 rs9621532 single nucleotide polymorphisms. Discov Oncol 2023; 14:8. [PMID: 36682035 PMCID: PMC9867797 DOI: 10.1007/s12672-023-00619-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/16/2023] [Indexed: 01/23/2023] Open
Abstract
PURPOSE Results of laryngeal squamous cell carcinoma (LSCC) treatment and the 5 year survival rate of these patients remain poor. To purify therapeutic targets, investigation of new specific and prognostic blood-based markers for LSCC development is essential. METHODS In the present study, we evaluated five single nucleotide polymorphisms (SNPs): IL1RAP rs4624606, IL1RL1 rs1041973, IL-6 rs1800795, BLK rs13277113, and TIMP3 rs9621532, and determined their associations with the patients' 5 year survival rate. Also, we performed a detailed statistical analysis of different LSCC patients' characteristics impact on their survival rate. RESULTS Three hundred fifty-three LSCC patients and 538 control subjects were included in this study. The multivariable Cox regression analysis revealed a significant association between patients' survival rate and distribution of IL1RAP rs4624606 variants: patients carrying AT genotype at IL1RAP rs4624606 had a lower risk of death (p = 0.044). Also, it was revealed that tumor size (T) (p = 0.000), tumor differentiation grade (G) (p = 0.015), and IL1RAP rs4624606 genotype (p = 0.044) were effective variables in multivariable Cox regression analysis prognosing survival of LSCC patients. The specific-LSCC 5 year survival rate was 77%. CONCLUSIONS In summary, our findings indicate that the genotypic distribution of IL1RAP rs4624606 influences the 5 year survival rate of LSCC patients. The results of the present study facilitate a more complete understanding of LSCC at the biological level, thus providing the base for the identification of new specific and prognostic blood-based markers for LSCC development.
Collapse
Affiliation(s)
- Agne Pasvenskaite
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Rasa Liutkeviciene
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), Kaunas, Lithuania
| | - Greta Gedvilaite
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), Kaunas, Lithuania
| | - Alvita Vilkeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences (LUHS), Kaunas, Lithuania
| | - Vykintas Liutkevicius
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| | - Virgilijus Uloza
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences (LUHS), A. Mickeviciaus 9, LT 44307 Kaunas, Lithuania
| |
Collapse
|
9
|
Choi S, Lee YJ, Choi Y, Kim M, Kim HJ, Kim JE, Oh S, Chae SW, Cha HJ, Jo JC. Prognostic significance of BLK expression in R-CHOP treated diffuse large B-cell lymphoma. J Pathol Transl Med 2022; 56:281-288. [PMID: 36128864 PMCID: PMC9510039 DOI: 10.4132/jptm.2022.07.26] [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: 04/07/2022] [Accepted: 07/26/2022] [Indexed: 11/24/2022] Open
Abstract
Background The aim of the present study was to evaluate the prognostic significance of B-cell lymphocyte kinase (BLK) expression for survival outcomes in diffuse large B-cell lymphoma (DLBCL) patients treated with R-CHOP. Methods We retrospectively analyzed the medical records of 89 patients from two tertiary referral hospitals. The expression of BLK, SYK, and CDK1 were evaluated in a semi-quantitative method using an H-score, and the proportions of BCL2 and C-MYC were evaluated. Results A total of 89 patients received R-CHOP chemotherapy as a first-line chemotherapy. The expression rates of BLK in tumor cells was 39.2% (n = 34). BLK expression status was not significantly associated with clinical variables; however, BLK expression in tumor cells was significantly associated with the expression of both C-MYC and BCL2 (p = .003). With a median follow-up of 60.4 months, patients with BLK expression had significantly lower 5-year progression-free survival (PFS) and overall survival rates (49.8% and 60.9%, respectively) than patients without BLK expression (77.3% and 86.7%, respectively). In multivariate analysis for PFS, BLK positivity was an independent poor prognostic factor (hazard ratio, 2.208; p = .040). Conclusions Here, we describe the clinicopathological features and survival outcome according to expression of BLK in DLBCL. Approximately 39% of DLBCL patients showed BLK positivity, which was associated as a predictive marker for poor prognosis in patients who received R-CHOP chemotherapy.
Collapse
Affiliation(s)
- Soyeon Choi
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Yoo Jin Lee
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Yunsuk Choi
- Department of Hematology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Misung Kim
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Hyun-Jung Kim
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Ji Eun Kim
- Department of Pathology, Seoul National University Boramae Hospital, Seoul, Korea
| | - Sukjoong Oh
- Department of Hematology and Oncology, Hanyang University Medical Center, Seoul, Korea
| | - Seoung Wan Chae
- Department of Pathology, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Hee Jeong Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Jae-Cheol Jo
- Department of Hematology and Oncology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| |
Collapse
|
10
|
Vinyals A, Ferreres JR, Calbet-Llopart N, Ramos R, Tell-Martí G, Carrera C, Marcoval J, Puig S, Malvehy J, Puig-Butillé JA, Fabra À. Oncogenic properties via MAPK signaling of the SOX5-RAF1 fusion gene identified in a wild-type NRAS/BRAF giant congenital nevus. Pigment Cell Melanoma Res 2022; 35:450-460. [PMID: 35587097 DOI: 10.1111/pcmr.13044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 04/07/2022] [Accepted: 05/02/2022] [Indexed: 01/17/2023]
Abstract
We recently reported an RAF rearrangement without NRAS or BRAF mutations in lesions from Giant Congenital Melanocytic Nevi (CMN). The new gene fusion involves the 5'-end of the promoter-containing N terminus of the SOX5 gene fused to exons 7-16 of the 3'-end of RAF1 gene leading to a SOX5-RAF1 fusion transcript which loses the auto-inhibitory CR1 domain but retains the complete in-frame coding sequence for the C-Terminal kinase domain of the RAF1. Stable expression of SOX5-RAF1 fusion induced growth factor-independent cell growth in murine hematopoietic Ba/F3 cells and melan-a immortalized melanocytes. Besides, it led to the transformation of both Ba/F3 and NIH 3T3 cells as revealed by colony formation assays. Furthermore, its expression results in MAPK activation assessed by increased levels of p-ERK protein in the cytosol of transduced cells. Treatment with Sorafenib and UO126 inhibited proliferation of Ba/F3-SOX5-RAF1 cells in the absence of IL3 but not the PLX 4720, a specific inhibitor of BRAF. Moreover, the tumorigenic and metastatic capacities of SOX5-RAF1 were assessed in vivo. These results indicate that SOX5-RAF1, a driver event for CMN development, has oncogenic capacity. Thus, sequencing of CMN transcriptomes may lead to the identification of this druggable fusion and interfere with the progression toward melanoma.
Collapse
Affiliation(s)
- Antònia Vinyals
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Josep R Ferreres
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain.,Dermatology Service, IDIBELL - Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Neus Calbet-Llopart
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Raquel Ramos
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Gemma Tell-Martí
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Cristina Carrera
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Joaquim Marcoval
- Dermatology Service, IDIBELL - Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Susana Puig
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Josep Malvehy
- Dermatology Department, IDIBAPS, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain
| | - Joan Anton Puig-Butillé
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Barcelona, Spain.,Molecular Biology CORE Laboratory, Melanoma Unit, IDIBAPS, Hospital Clínic de Barcelona, University of Barcelona, Barcelona, Spain
| | - Àngels Fabra
- Oncobell Program, Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| |
Collapse
|
11
|
Chai H, Zhou X, Zhang Z, Rao J, Zhao H, Yang Y. Integrating multi-omics data through deep learning for accurate cancer prognosis prediction. Comput Biol Med 2021; 134:104481. [PMID: 33989895 DOI: 10.1016/j.compbiomed.2021.104481] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 05/06/2021] [Accepted: 05/06/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Genomic information is nowadays widely used for precise cancer treatments. Since the individual type of omics data only represents a single view that suffers from data noise and bias, multiple types of omics data are required for accurate cancer prognosis prediction. However, it is challenging to effectively integrate multi-omics data due to the large number of redundant variables but relatively small sample size. With the recent progress in deep learning techniques, Autoencoder was used to integrate multi-omics data for extracting representative features. Nevertheless, the generated model is fragile from data noises. Additionally, previous studies usually focused on individual cancer types without making comprehensive tests on pan-cancer. Here, we employed the denoising Autoencoder to get a robust representation of the multi-omics data, and then used the learned representative features to estimate patients' risks. RESULTS By applying to 15 cancers from The Cancer Genome Atlas (TCGA), our method was shown to improve the C-index values over previous methods by 6.5% on average. Considering the difficulty to obtain multi-omics data in practice, we further used only mRNA data to fit the estimated risks by training XGboost models, and found the models could achieve an average C-index value of 0.627. As a case study, the breast cancer prognosis prediction model was independently tested on three datasets from the Gene Expression Omnibus (GEO), and shown able to significantly separate high-risk patients from low-risk ones (C-index>0.6, p-values<0.05). Based on the risk subgroups divided by our method, we identified nine prognostic markers highly associated with breast cancer, among which seven genes have been proved by literature review. CONCLUSION Our comprehensive tests indicated that we have constructed an accurate and robust framework to integrate multi-omics data for cancer prognosis prediction. Moreover, it is an effective way to discover cancer prognosis-related genes.
Collapse
Affiliation(s)
- Hua Chai
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Xiang Zhou
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Zhongyue Zhang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Jiahua Rao
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China
| | - Huiying Zhao
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510000, China.
| | - Yuedong Yang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, 510000, China; Key Laboratory of Machine Intelligence and Advanced Computing (MOE), Sun Yat-sen University, Guangzhou, 510000, China.
| |
Collapse
|
12
|
Zhai Y, Yang J, Zhang J, Yang J, Li Q, Zheng T. Src-family Protein Tyrosine Kinases: A promising target for treating Cardiovascular Diseases. Int J Med Sci 2021; 18:1216-1224. [PMID: 33526983 PMCID: PMC7847615 DOI: 10.7150/ijms.49241] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/18/2020] [Indexed: 02/07/2023] Open
Abstract
The Src-family protein tyrosine kinases (SFKs), a subfamily of non-receptor tyrosine kinases, are ubiquitously expressed in various cell types. Numerous studies have suggested that SFKs are related to signal transduction in major cardiac physiological and pathological processes, it is the activity of SFKs that is connected with the maintenance of cardiovascular homeostasis. Upon stimulation of various injury factors or stress, the phosphorylation state of SFKs is changed, which has been found to modulate different cardiac pathological conditions, such as hypertension, coronary heart disease, ischemic heart disease, myocardial ischemia-reperfusion injury, arrhythmia and cardiomyopathy via regulating cell growth, differentiation, movement and function, electrophysiologic signals. This review summarizes the basic information about SFKs, updates its role in the different processes underlying the development of multiple cardiovascular diseases (CVDs), and highlights their potential role as disease biomarkers and therapeutic targets, which would help understand the pathophysiology of CVDs and promote the further potential clinical adhibition.
Collapse
Affiliation(s)
- Yuhong Zhai
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
- Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| | - Jun Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
| | - Jing Zhang
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
- Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China
| | - Qi Li
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
- Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| | - Tao Zheng
- Department of Cardiology, The First College of Clinical Medical Science, China Three Gorges University, Yichang 443000, China
- Institute of Cardiovascular Diseases, China Three Gorges University, Yichang 443000, China
- Central Laboratory, Yichang Central People's Hospital, Yichang 443000, China
| |
Collapse
|
13
|
Creeden JF, Alganem K, Imami AS, Henkel ND, Brunicardi FC, Liu SH, Shukla R, Tomar T, Naji F, McCullumsmith RE. Emerging Kinase Therapeutic Targets in Pancreatic Ductal Adenocarcinoma and Pancreatic Cancer Desmoplasia. Int J Mol Sci 2020; 21:ijms21228823. [PMID: 33233470 PMCID: PMC7700673 DOI: 10.3390/ijms21228823] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/16/2020] [Accepted: 11/19/2020] [Indexed: 02/08/2023] Open
Abstract
Kinase drug discovery represents an active area of therapeutic research, with previous pharmaceutical success improving patient outcomes across a wide variety of human diseases. In pancreatic ductal adenocarcinoma (PDAC), innovative pharmaceutical strategies such as kinase targeting have been unable to appreciably increase patient survival. This may be due, in part, to unchecked desmoplastic reactions to pancreatic tumors. Desmoplastic stroma enhances tumor development and progression while simultaneously restricting drug delivery to the tumor cells it protects. Emerging evidence indicates that many of the pathologic fibrotic processes directly or indirectly supporting desmoplasia may be driven by targetable protein tyrosine kinases such as Fyn-related kinase (FRK); B lymphoid kinase (BLK); hemopoietic cell kinase (HCK); ABL proto-oncogene 2 kinase (ABL2); discoidin domain receptor 1 kinase (DDR1); Lck/Yes-related novel kinase (LYN); ephrin receptor A8 kinase (EPHA8); FYN proto-oncogene kinase (FYN); lymphocyte cell-specific kinase (LCK); tec protein kinase (TEC). Herein, we review literature related to these kinases and posit signaling networks, mechanisms, and biochemical relationships by which this group may contribute to PDAC tumor growth and desmoplasia.
Collapse
Affiliation(s)
- Justin F. Creeden
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (K.A.); (A.S.I.); (N.D.H.); (R.S.); (R.E.M.)
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (F.C.B.); (S.-H.L.)
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 6038, USA
- Correspondence: ; Tel.: +1-419-383-6474
| | - Khaled Alganem
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (K.A.); (A.S.I.); (N.D.H.); (R.S.); (R.E.M.)
| | - Ali S. Imami
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (K.A.); (A.S.I.); (N.D.H.); (R.S.); (R.E.M.)
| | - Nicholas D. Henkel
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (K.A.); (A.S.I.); (N.D.H.); (R.S.); (R.E.M.)
| | - F. Charles Brunicardi
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (F.C.B.); (S.-H.L.)
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 6038, USA
| | - Shi-He Liu
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (F.C.B.); (S.-H.L.)
- Department of Surgery, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 6038, USA
| | - Rammohan Shukla
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (K.A.); (A.S.I.); (N.D.H.); (R.S.); (R.E.M.)
| | - Tushar Tomar
- PamGene International BV, 5200 BJ’s-Hertogenbosch, The Netherlands; (T.T.); (F.N.)
| | - Faris Naji
- PamGene International BV, 5200 BJ’s-Hertogenbosch, The Netherlands; (T.T.); (F.N.)
| | - Robert E. McCullumsmith
- Department of Neurosciences, College of Medicine and Life Sciences, University of Toledo, Toledo, OH 43614, USA; (K.A.); (A.S.I.); (N.D.H.); (R.S.); (R.E.M.)
- Neurosciences Institute, ProMedica, Toledo, OH 6038, USA
| |
Collapse
|
14
|
Behravan H, Hartikainen JM, Tengström M, Kosma VM, Mannermaa A. Predicting breast cancer risk using interacting genetic and demographic factors and machine learning. Sci Rep 2020; 10:11044. [PMID: 32632202 PMCID: PMC7338351 DOI: 10.1038/s41598-020-66907-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2020] [Accepted: 06/01/2020] [Indexed: 12/21/2022] Open
Abstract
Breast cancer (BC) is a multifactorial disease and the most common cancer in women worldwide. We describe a machine learning approach to identify a combination of interacting genetic variants (SNPs) and demographic risk factors for BC, especially factors related to both familial history (Group 1) and oestrogen metabolism (Group 2), for predicting BC risk. This approach identifies the best combinations of interacting genetic and demographic risk factors that yield the highest BC risk prediction accuracy. In tests on the Kuopio Breast Cancer Project (KBCP) dataset, our approach achieves a mean average precision (mAP) of 77.78 in predicting BC risk by using interacting genetic and Group 1 features, which is better than the mAPs of 74.19 and 73.65 achieved using only Group 1 features and interacting SNPs, respectively. Similarly, using interacting genetic and Group 2 features yields a mAP of 78.00, which outperforms the system based on only Group 2 features, which has a mAP of 72.57. Furthermore, the gene interaction maps built from genes associated with SNPs that interact with demographic risk factors indicate important BC-related biological entities, such as angiogenesis, apoptosis and oestrogen-related networks. The results also show that demographic risk factors are individually more important than genetic variants in predicting BC risk.
Collapse
Affiliation(s)
- Hamid Behravan
- Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland.
| | - Jaana M Hartikainen
- Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
| | - Maria Tengström
- Institute of Clinical Medicine, Oncology, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
- Cancer Center, Kuopio University Hospital, Kuopio, P.O. Box 100, FI-70029, Kuopio, Finland
| | - Veli-Matti Kosma
- Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| | - Arto Mannermaa
- Institute of Clinical Medicine, Pathology and Forensic Medicine, and Translational Cancer Research Area, University of Eastern Finland, P.O. Box 1627, FI-70211, Kuopio, Finland
- Biobank of Eastern Finland, Kuopio University Hospital, Kuopio, Finland
| |
Collapse
|
15
|
Pasvenskaite A, Vilkeviciute A, Liutkeviciene R, Gedvilaite G, Liutkevicius V, Uloza V. Associations of IL6 rs1800795, BLK rs13277113, TIMP3 rs9621532, IL1RL1 rs1041973 and IL1RAP rs4624606 single gene polymorphisms with laryngeal squamous cell carcinoma. Gene 2020; 747:144700. [PMID: 32330537 DOI: 10.1016/j.gene.2020.144700] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2020] [Revised: 04/06/2020] [Accepted: 04/18/2020] [Indexed: 02/07/2023]
Abstract
OBJECTIVE Survival rate of laryngeal squamous cell carcinoma (LSCC) patients is not improving. To understand more complete biology of LSCC, studies focused on identification of new specific and prognostic markers are performed. The aim of current study was to evaluate the impact of five different single nucleotide polymorphisms (SNP) (IL6 rs1800795, BLK rs13277113, TIMP3 rs9621532, IL1RL1 rs1041973 and IL1RAP rs4624606) on LSCC development. MATERIAL AND METHODS A total of 891 subjects (353 histologically verified LSCC patients and 538 healthy controls) were involved in this study. The genotyping was carried out using the real-time-PCR. RESULTS Statistical analysis revealed statistically significant associations between TIMP3 rs96215332 variants and LSCC in the codominant (OR = 0.600; 95% CI: 0.390-0.922; p = 0.020), overdominant (OR = 0.599; 95% CI: 0.390-0.922; p = 0.020) and additive (OR = 0.675; 95% CI: 0.459-0.991; p = 0.045) models. Also, significant variants of IL1RAP rs4624606 were determined in the codominant (OR = 1.372; 95% CI: 1.031-1.827; p = 0.030), overdominant (OR = 1.353; 95% CI: 1.018-1.798; p = 0.037) and additive (OR = 1.337; 95% CI: 1.038-1.724; p = 0.025) models. CONCLUSION Results of the current study indicate significant associations between TIMP3 rs9621532 and IL1RAP rs4624606 gene polymorphisms and LSCC development.
Collapse
Affiliation(s)
- Agne Pasvenskaite
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences, Kaunas, Lithuania.
| | - Alvita Vilkeviciute
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania, Lithuania
| | - Rasa Liutkeviciene
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania, Lithuania
| | - Greta Gedvilaite
- Neuroscience Institute, Lithuanian University of Health Sciences, Kaunas, Lithuania, Lithuania
| | - Vykintas Liutkevicius
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Virgilijus Uloza
- Department of Otorhinolaryngology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| |
Collapse
|
16
|
Bagnato G, Leopizzi M, Urciuoli E, Peruzzi B. Nuclear Functions of the Tyrosine Kinase Src. Int J Mol Sci 2020; 21:ijms21082675. [PMID: 32290470 PMCID: PMC7215861 DOI: 10.3390/ijms21082675] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022] Open
Abstract
Src is the representative member of the Src-family kinases (SFKs), a group of tyrosine kinases involved in several cellular processes. Its main function has been for long confined to the plasma membrane/cytoplasm compartment, being a myristoylated protein anchored to the cell membrane and functioning downstream to receptors, most of them lacking intrinsic kinase activity. In the last decades, new roles for some SFKs have been described in the nuclear compartment, suggesting that these proteins can also be involved in directly regulating gene transcription or nucleoskeleton architecture. In this review, we focused on those nuclear functions specifically attributable to Src, by considering its function as both tyrosine kinase and adapting molecule. In particular, we addressed the Src involvement in physiological as well as in pathological conditions, especially in tumors.
Collapse
Affiliation(s)
- Giulia Bagnato
- Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.B.); (E.U.)
| | - Martina Leopizzi
- Department of Medico-Surgical Sciences and Biotechnology, Polo Pontino, Sapienza University, 04100 Latina, Italy;
| | - Enrica Urciuoli
- Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.B.); (E.U.)
| | - Barbara Peruzzi
- Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (G.B.); (E.U.)
- Correspondence:
| |
Collapse
|
17
|
Identification of recurrent noncoding mutations in B-cell lymphoma using capture Hi-C. Blood Adv 2020; 3:21-32. [PMID: 30606723 DOI: 10.1182/bloodadvances.2018026419] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 11/24/2018] [Indexed: 12/22/2022] Open
Abstract
The identification of driver mutations is fundamental to understanding oncogenesis. Although genes frequently mutated in B-cell lymphoma have been identified, the search for driver mutations has largely focused on the coding genome. Here we report an analysis of the noncoding genome using whole-genome sequencing data from 117 patients with B-cell lymphoma. Using promoter capture Hi-C data in naive B cells, we define cis-regulatory elements, which represent an enriched subset of the noncoding genome in which to search for driver mutations. Regulatory regions were identified whose mutation significantly alters gene expression, including copy number variation at cis-regulatory elements targeting CD69, IGLL5, and MMP14, and single nucleotide variants in a cis-regulatory element for TPRG1 We also show the commonality of pathways targeted by coding and noncoding mutations, exemplified by MMP14, which regulates Notch signaling, a pathway important in lymphomagenesis and whose expression is associated with patient survival. This study provides an enhanced understanding of lymphomagenesis and describes the advantages of using chromosome conformation capture to decipher noncoding mutations relevant to cancer biology.
Collapse
|
18
|
Soh PXY, Marin Cely JM, Mortlock SA, Jara CJ, Booth R, Natera S, Roessner U, Crossett B, Cordwell S, Singh Khatkar M, Williamson P. Genome-wide association studies of 74 plasma metabolites of German shepherd dogs reveal two metabolites associated with genes encoding their enzymes. Metabolomics 2019; 15:123. [PMID: 31493001 DOI: 10.1007/s11306-019-1586-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 08/29/2019] [Indexed: 12/12/2022]
Abstract
INTRODUCTION German shepherd dogs (GSDs) are a popular breed affected by numerous disorders. Few studies have explored genetic variations that influence canine blood metabolite levels. OBJECTIVES To investigate genetic variants affecting the natural metabolite variation in GSDs. METHODS A total of 82 healthy GSDs were genotyped on the Illumina CanineHD Beadchip, assaying 173,650 markers. For each dog, 74 metabolites were measured through liquid and gas chromatography mass spectrometry (LC-MS and GC-MS) and were used as phenotypes for genome-wide association analyses (GWAS). Sliding window and homozygosity analyses were conducted to fine-map regions of interest, and to identify haplotypes and gene dosage effects. RESULTS Summary statistics for 74 metabolites in this population of GSDs are reported. Forty-one metabolites had significant associations at a false discovery rate of 0.05. Two associations were located around genes which encode for enzymes for the relevant metabolites: 4-hydroxyproline was significantly associated to D-amino acid oxidase (DAO), and threonine to L-threonine 3-dehydrogenase (LOC477365). Three of the top ten haplotypes associated to 4-hydroxyproline included at least one SNP on DAO. These haplotypes occurred only in dogs with the highest 15 measurements of 4-hydroxyproline, ranging in frequency from 16.67 to 20%. None of the dogs were homozygous for these haplotypes. The top two haplotypes associated to threonine included SNPs on LOC477365 and were also overrepresented in dogs with the highest 15 measurements of threonine. These haplotypes occurred at a frequency of 90%, with 80% of these dogs homozygous for the haplotypes. In dogs with the lowest 15 measurements of threonine, the haplotypes occurred at a frequency of 26.67% and 0% homozygosity. CONCLUSION DAO and LOC477365 were identified as candidate genes affecting the natural plasma concentration of 4-hydroxyproline and threonine, respectively. Further investigations are needed to validate the effects of the variants on these genes.
Collapse
Affiliation(s)
- Pamela Xing Yi Soh
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia
| | - Juliana Maria Marin Cely
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia
| | - Sally-Anne Mortlock
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia
| | - Christopher James Jara
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia
| | - Rachel Booth
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia
| | - Siria Natera
- Metabolomics Australia, School of BioSciences, University of Melbourne, Parkville, Australia
| | - Ute Roessner
- Metabolomics Australia, School of BioSciences, University of Melbourne, Parkville, Australia
| | - Ben Crossett
- Sydney Mass Spectrometry, Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Stuart Cordwell
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia
- Sydney Mass Spectrometry, Charles Perkins Centre, University of Sydney, Sydney, Australia
| | - Mehar Singh Khatkar
- Sydney School of Veterinary Science, Faculty of Science, University of Sydney, Sydney, Australia
| | - Peter Williamson
- School of Life and Environmental Sciences, Faculty of Science, University of Sydney, Sydney, Australia.
| |
Collapse
|
19
|
Solouki S, August A, Huang W. Non-receptor tyrosine kinase signaling in autoimmunity and therapeutic implications. Pharmacol Ther 2019; 201:39-50. [PMID: 31082431 DOI: 10.1016/j.pharmthera.2019.05.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 04/18/2019] [Indexed: 12/17/2022]
Abstract
Autoimmune diseases are characterized by impaired immune tolerance towards self-antigens, leading to enhanced immunity to self by dysfunctional B cells and/or T cells. The activation of these cells is controlled by non-receptor tyrosine kinases (NRTKs), which are critical mediators of antigen receptor and cytokine receptor signaling pathways. NRTKs transduce, amplify and sustain activating signals that contribute to autoimmunity, and are counter-regulated by protein tyrosine phosphatases (PTPs). The function of and interaction between NRTKs and PTPs during the development of autoimmunity could be key points of therapeutic interference against autoimmune diseases. In this review, we summarize the current state of knowledge of the functions of NRTKs and PTPs involved in B cell receptor (BCR), T cell receptor (TCR), and cytokine receptor signaling pathways that contribute to autoimmunity, and discuss their targeting for therapeutic approaches against autoimmune diseases.
Collapse
Affiliation(s)
- Sabrina Solouki
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Avery August
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA.
| | - Weishan Huang
- Department of Microbiology and Immunology, College of Veterinary Medicine, Cornell University, Ithaca, NY 14853, USA; Department of Pathobiological Sciences, School of Veterinary Medicine, Louisiana State University, Baton Rouge, LA 70803, USA.
| |
Collapse
|
20
|
Liu H, Zhang H, Wu X, Ma D, Wu J, Wang L, Jiang Y, Fei Y, Zhu C, Tan R, Jungblut P, Pei G, Dorhoi A, Yan Q, Zhang F, Zheng R, Liu S, Liang H, Liu Z, Yang H, Chen J, Wang P, Tang T, Peng W, Hu Z, Xu Z, Huang X, Wang J, Li H, Zhou Y, Liu F, Yan D, Kaufmann SHE, Chen C, Mao Z, Ge B. Nuclear cGAS suppresses DNA repair and promotes tumorigenesis. Nature 2018; 563:131-136. [PMID: 30356214 DOI: 10.1038/s41586-018-0629-6] [Citation(s) in RCA: 458] [Impact Index Per Article: 65.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2017] [Accepted: 08/22/2018] [Indexed: 12/24/2022]
Abstract
Accurate repair of DNA double-stranded breaks by homologous recombination preserves genome integrity and inhibits tumorigenesis. Cyclic GMP-AMP synthase (cGAS) is a cytosolic DNA sensor that activates innate immunity by initiating the STING-IRF3-type I IFN signalling cascade1,2. Recognition of ruptured micronuclei by cGAS links genome instability to the innate immune response3,4, but the potential involvement of cGAS in DNA repair remains unknown. Here we demonstrate that cGAS inhibits homologous recombination in mouse and human models. DNA damage induces nuclear translocation of cGAS in a manner that is dependent on importin-α, and the phosphorylation of cGAS at tyrosine 215-mediated by B-lymphoid tyrosine kinase-facilitates the cytosolic retention of cGAS. In the nucleus, cGAS is recruited to double-stranded breaks and interacts with PARP1 via poly(ADP-ribose). The cGAS-PARP1 interaction impedes the formation of the PARP1-Timeless complex, and thereby suppresses homologous recombination. We show that knockdown of cGAS suppresses DNA damage and inhibits tumour growth both in vitro and in vivo. We conclude that nuclear cGAS suppresses homologous-recombination-mediated repair and promotes tumour growth, and that cGAS therefore represents a potential target for cancer prevention and therapy.
Collapse
Affiliation(s)
- Haipeng Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haiping Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiangyang Wu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dapeng Ma
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Juehui Wu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lin Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yan Jiang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiyan Fei
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai, China
| | - Chenggang Zhu
- Department of Optical Science and Engineering, Shanghai Engineering Research Center of Ultra-Precision Optical Manufacturing, Key Laboratory of Micro and Nano Photonic Structures (Ministry of Education), Fudan University, Shanghai, China
| | - Rong Tan
- Center for Molecular Medicine, Xiangya Hospital, Central South University, Changsha, China
| | - Peter Jungblut
- Protein Purification Core Facility, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Gang Pei
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Anca Dorhoi
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
- Institute of Immunology, Friedrich-Loeffler-Institut, Federal Research Institute for Animal Health, Greifswald Insel Riems, Germany
| | - Qiaoling Yan
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fan Zhang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ruijuan Zheng
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Siyu Liu
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haijiao Liang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhonghua Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Hua Yang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jianxia Chen
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Peng Wang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianqi Tang
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Wenxia Peng
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhangsen Hu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Zhu Xu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaochen Huang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jie Wang
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haohao Li
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yilong Zhou
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Liu
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Dapeng Yan
- Key Laboratory of Medical Molecular Virology of MOE/MOH, Fudan University, Shanghai, China
| | - Stefan H E Kaufmann
- Department of Immunology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhiyong Mao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.
| | - Baoxue Ge
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
- Clinical Translational Research Center, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|