1
|
Liu X, Shi X, Zhao H, Wang C. Exploring the molecular mechanisms of comorbidity of myocardial infarction and anxiety disorders by combining multiple data sets with in vivo experimental validation. Int Immunopharmacol 2025; 146:113852. [PMID: 39733641 DOI: 10.1016/j.intimp.2024.113852] [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: 09/24/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 12/31/2024]
Abstract
BACKGROUND The incidence of comorbidity between myocardial infarction (MI) and anxiety disorders is increasing. However, the biological association between them has not been fully understood. OBJECTIVE This study aims to investigate the molecular mechanisms of comorbidity between MI and anxiety disorders and to predict their key genes and potential therapeutic drugs. METHODS We searched Gene Expression Omnibus databases and performed differential analyses using the limma package to identify the functional enrichment of differential genes. Next, we constructed regulatory networks to investigate the relationship between hub genes and autophagy, ferroptosis, and immunity. Furthermore, we predicted transcription factors by R package, constructed a miRNA network, performed the single-cell analysis of key gene expression, and predicted drug targeting of differential genes using the Connectivity Map database. RESULTS The datasets for MI and anxiety disorders were analyzed for up and down-regulated differential genes, resulting in 35 intersecting differential genes. The top 10 feature genes from each dataset were intersected using Random Forest, resulting in the identification of three intersecting genes: STK17B, AKIRIN2, and WDR77. Validation of the above key genes was carried out by in vitro experiments. We examined the gene expression of STK17B, WDR77 and AKIRIN2 in the hippocampus and myocardial infarction border zone respectively by qPCR and WB, and the results confirmed that the above are the key genes for myocardial infarction and anxiety. There is a significant correlation between the comorbidity mechanism of myocardial infarction and anxiety disorders with ferroptosis and immunity. The construction of the miRNA network revealed that miR-205 and let-7 had higher average connectivity among the three hub genes. The single-cell analysis revealed significant expression of key genes in Endothelial cells, Cardiomyocytes, Macrophages, and Fibroblasts datasets. Cd274 showed a higher correlation with key genes in myocardial infarction and anxiety disorders. CONCLUSION Validation by multiple datasets and in vitro experiments showed that STK17B, AKIRIN2, and WDR77 are the key genes in the comorbidity of myocardial infarction and anxiety disorders, and ferroptosis and immunity are the key links in the comorbidity mechanism of myocardial infarction and anxiety disorders.
Collapse
Affiliation(s)
- Xiang Liu
- Beijing University of Chinese Medicine, Beijing, China.
| | - Xiaojun Shi
- Beijing University of Chinese Medicine, Beijing, China
| | - Haibin Zhao
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China
| | - Chao Wang
- Dongfang Hospital of Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
2
|
Sun Z, Hu M, Huang X, Song M, Chen X, Bei J, Lin Y, Chen S. Predictive value of dendritic cell-related genes for prognosis and immunotherapy response in lung adenocarcinoma. Cancer Cell Int 2025; 25:13. [PMID: 39810206 PMCID: PMC11730157 DOI: 10.1186/s12935-025-03642-z] [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: 07/25/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Patients with lung adenocarcinoma (LUAD) receiving drug treatment often have an unpredictive response and there is a lack of effective methods to predict treatment outcome for patients. Dendritic cells (DCs) play a significant role in the tumor microenvironment and the DCs-related gene signature may be used to predict treatment outcome. Here, we screened for DC-related genes to construct a prognostic signature to predict prognosis and response to immunotherapy in LUAD patients. METHODS DC-related biological functions and genes were identified using single-cell RNA sequencing (scRNA-seq) and bulk RNA sequencing. DCs-related gene signature (DCRGS) was constructed using integrated machine learning algorithms. Expression of key genes in clinical samples was examined by real-time q-PCR. Performance of the prognostic model, DCRGS, for the prognostic evaluation, was assessed using a multiple time-dependent receiver operating characteristic (ROC) curve, the R package, "timeROC", and validated using GEO datasets. RESULTS Analysis of scRNA-seq data showed that there is a significant upregulation of LGALS9 expression in DCs isolated from malignant pleural effusion samples. Leveraging the Coxboost and random survival forest combination algorithm, we filtered out six DC-related genes on which a prognostic prediction model, DCRGS, was established. A high predictive capability nomogram was constructed by combining DCRGS with clinical features. We found that patients with a high-DCRGS score had immunosuppression, activated tumor-associated pathways, and elevated somatic mutational load and copy number variant load. In contrast, patients in the low-DCRGS subgroup were resistant to chemotherapy but sensitive to the CTLA-4 immune checkpoint inhibitor and targeted therapy. CONCLUSION We have innovatively established a deep learning-based prediction model, DCRGS, for the prediction of the prognosis of patients with LUAD. The model possesses a strong prognostic prediction performance with high accuracy and sensitivity and could be clinically useful to guide the management of LUAD. Furthermore, the findings of this study could provide an important reference for individualized clinical treatment and prognostic prediction of patients with LUAD.
Collapse
Affiliation(s)
- Zihao Sun
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Mengfei Hu
- Department of Internal Medicine, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, 230000, China
| | - Xiaoning Huang
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Minghan Song
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Xiujing Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Jiaxin Bei
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
| | - Yiguang Lin
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Research & Development Division, Guangzhou Anjie Biomedical Technology Co., Ltd., Guangzhou, 510535, China.
| | - Size Chen
- Department of Immuno-Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Guangdong Provincial Engineering Research Center for Esophageal Cancer Precision Therapy, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China.
- Key Laboratory of Cancer Immunotherapy of Guangdong Higher Education Institutes, Guangzhou, 510080, China.
- Key Laboratory of Monitoring Adverse Reactions Associated with CAR-T Cell Therapy, Guangzhou, 510080, China.
| |
Collapse
|
3
|
Li C, Weng J, Yang L, Gong H, Liu Z. Development of an anoikis-related gene signature and prognostic model for predicting the tumor microenvironment and response to immunotherapy in colorectal cancer. Front Immunol 2024; 15:1378305. [PMID: 38779664 PMCID: PMC11109372 DOI: 10.3389/fimmu.2024.1378305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024] Open
Abstract
The effect of anoikis-related genes (ARGs) on clinicopathological characteristics and tumor microenvironment remains unclear. We comprehensively analyzed anoikis-associated gene signatures of 1057 colorectal cancer (CRC) samples based on 18 ARGs. Anoikis-related molecular subtypes and gene features were identified through consensus clustering analysis. The biological functions and immune cell infiltration were assessed using the GSVA and ssGSEA algorithms. Prognostic risk score was constructed using multivariate Cox regression analysis. The immunological features of high-risk and low-risk groups were compared. Finally, DAPK2-overexpressing plasmid was transfected to measure its effect on tumor proliferation and metastasis in vitro and in vivo. We identified 18 prognostic ARGs. Three different subtypes of anoikis were identified and demonstrated to be linked to distinct biological processes and prognosis. Then, a risk score model was constructed and identified as an independent prognostic factor. Compared to the high-risk group, patients in the low-risk group exhibited longer survival, higher enrichment of checkpoint function, increased expression of CTLA4 and PD-L1, higher IPS scores, and a higher proportion of MSI-H. The results of RT-PCR indicated that the expression of DAPK2 mRNA was significantly downregulated in CRC tissues compared to normal tissues. Increased DAPK2 expression significantly suppressed cell proliferation, promoted apoptosis, and inhibited migration and invasion. The nude mice xenograft tumor model confirmed that high expression of DAPK2 inhibited tumor growth. Collectively, we discovered an innovative anoikis-related gene signature associated with prognosis and TME. Besides, our study indicated that DAPK2 can serve as a promising therapeutic target for inhibiting the growth and metastasis of CRC.
Collapse
Affiliation(s)
- Chuanchang Li
- Department of General Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Junyong Weng
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Le Yang
- Department of General Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hangjun Gong
- Department of Gastrointestinal Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaolong Liu
- Department of General Surgery, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
4
|
Almeida A, T'Sas S, Pagliaro L, Fijalkowski I, Sleeckx W, Van Steenberge H, Zamponi R, Lintermans B, Van Loocke W, Palhais B, Reekmans A, Bardelli V, Demoen L, Reunes L, Deforce D, Van Nieuwerburgh F, Kentsis A, Ntziachristos P, Van Roy N, De Moerloose B, Mecucci C, La Starza R, Roti G, Goossens S, Van Vlierberghe P, Pieters T. Myb overexpression synergizes with the loss of Pten and is a dependency factor and therapeutic target in T-cell lymphoblastic leukemia. Hemasphere 2024; 8:e51. [PMID: 38463444 PMCID: PMC10924755 DOI: 10.1002/hem3.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 01/28/2024] [Indexed: 03/12/2024] Open
Abstract
T-lineage acute lymphoblastic leukemia (T-ALL) is an aggressive hematological malignancy that accounts for 10%-15% of pediatric and 25% of adult ALL cases. Although the prognosis of T-ALL has improved over time, the outcome of T-ALL patients with primary resistant or relapsed leukemia remains poor. Therefore, further progress in the treatment of T-ALL requires a better understanding of its biology and the development of more effective precision oncologic therapies. The proto-oncogene MYB is highly expressed in diverse hematologic malignancies, including T-ALLs with genomic aberrations that further potentiate its expression and activity. Previous studies have associated MYB with a malignant role in the pathogenesis of several cancers. However, its role in the induction and maintenance of T-ALL remains relatively poorly understood. In this study, we found that an increased copy number of MYB is associated with higher MYB expression levels, and might be associated with inferior event-free survival of pediatric T-ALL patients. Using our previously described conditional Myb overexpression mice, we generated two distinct MYB-driven T-ALL mouse models. We demonstrated that the overexpression of Myb synergizes with Pten deletion but not with the overexpression of Lmo2 to accelerate the development of T-cell lymphoblastic leukemias. We also showed that MYB is a dependency factor in T-ALL since RNA interference of Myb blocked cell cycle progression and induced apoptosis in both human and murine T-ALL cell lines. Finally, we provide preclinical evidence that targeting the transcriptional activity of MYB can be a useful therapeutic strategy for the treatment of T-ALL.
Collapse
Affiliation(s)
- André Almeida
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Sara T'Sas
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Luca Pagliaro
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Igor Fijalkowski
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Wouter Sleeckx
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Hannah Van Steenberge
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | | | - Béatrice Lintermans
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Wouter Van Loocke
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Bruno Palhais
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Alexandra Reekmans
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Valentina Bardelli
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Lisa Demoen
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Lindy Reunes
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Dieter Deforce
- Laboratory of Pharmaceutical BiotechnologyGhent UniversityGhentBelgium
| | | | - Alex Kentsis
- Tow Center for Developmental Oncology, Sloan Kettering Institute and Department of PediatricsMemorial Sloan Kettering Cancer CenterNew YorkNew YorkUSA
| | - Panagiotis Ntziachristos
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Nadine Van Roy
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Lab for Translational Oncogenomics and Bioinformatics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Pediatric Precision Oncology Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| | - Barbara De Moerloose
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Department of Pediatric Hematology‐OncologyGhent University HospitalGhentBelgium
| | - Cristina Mecucci
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Roberta La Starza
- Institute of Hematology and Center for Hemato‐Oncology ResearchUniversity of Perugia and S.M. Misericordia HospitalPerugiaItaly
| | - Giovanni Roti
- Department of Medicine and SurgeryUniversity of ParmaParmaItaly
| | - Steven Goossens
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
| | - Pieter Van Vlierberghe
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
| | - Tim Pieters
- Normal and Malignant Hematopoiesis Lab, Department of Biomolecular MedicineGhent UniversityGhentBelgium
- Cancer Research Institute Ghent (CRIG)GhentBelgium
- Unit for Translational Research in Oncology, Department of Diagnostic SciencesGhent UniversityGhentBelgium
- Leukemia Therapy Resistance Laboratory and Center for Medical Genetics, Department of Biomolecular MedicineGhent UniversityGhentBelgium
| |
Collapse
|
5
|
Anand S, Vikramdeo KS, Sudan SK, Sharma A, Acharya S, Khan MA, Singh S, Singh AP. From modulation of cellular plasticity to potentiation of therapeutic resistance: new and emerging roles of MYB transcription factors in human malignancies. Cancer Metastasis Rev 2024; 43:409-421. [PMID: 37950087 PMCID: PMC11015973 DOI: 10.1007/s10555-023-10153-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/05/2023] [Indexed: 11/12/2023]
Abstract
MYB transcription factors are encoded by a large family of highly conserved genes from plants to vertebrates. There are three members of the MYB gene family in human, namely, MYB, MYBL1, and MYBL2 that encode MYB/c-MYB, MYBL1/A-MYB, and MYBL2/B-MYB, respectively. MYB was the first member to be identified as a cellular homolog of the v-myb oncogene carried by the avian myeloblastosis virus (AMV) causing leukemia in chickens. Under the normal scenario, MYB is predominantly expressed in hematopoietic tissues, colonic crypts, and neural stem cells and plays a role in maintaining the undifferentiated state of the cells. Over the years, aberrant expression of MYB genes has been reported in several malignancies and recent years have witnessed tremendous progress in understanding of their roles in processes associated with cancer development. Here, we review various MYB alterations reported in cancer along with the roles of MYB family proteins in tumor cell plasticity, therapy resistance, and other hallmarks of cancer. We also discuss studies that provide mechanistic insights into the oncogenic functions of MYB transcription factors to identify potential therapeutic vulnerabilities.
Collapse
Affiliation(s)
- Shashi Anand
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Kunwar Somesh Vikramdeo
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Sarabjeet Kour Sudan
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Amod Sharma
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Srijan Acharya
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Mohammad Aslam Khan
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
| | - Seema Singh
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA
- Department of Biochemistry and Molecular Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36688, USA
| | - Ajay Pratap Singh
- Department of Pathology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36617, USA.
- Mitchell Cancer Institute, University of South Alabama, Mobile, AL, 36604, USA.
- Department of Biochemistry and Molecular Biology, Frederick P. Whiddon College of Medicine, University of South Alabama, Mobile, AL, 36688, USA.
| |
Collapse
|
6
|
Zheng Y, Li X, Kuang L, Wang Y. New insights into the characteristics of DRAK2 and its role in apoptosis: From molecular mechanisms to clinically applied potential. Front Pharmacol 2022; 13:1014508. [PMID: 36386181 PMCID: PMC9649744 DOI: 10.3389/fphar.2022.1014508] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 10/12/2022] [Indexed: 11/27/2022] Open
Abstract
As a member of the death-associated protein kinase (DAPK) family, DAP kinase-associated apoptosis-inducing kinase 2 (DRAK2) performs apoptosis-related functions. Compelling evidence suggests that DRAK2 is involved in regulating the activation of T lymphocytes as well as pancreatic β-cell apoptosis in type I diabetes. In addition, DRAK2 has been shown to be involved in the development of related tumor and non-tumor diseases through a variety of mechanisms, including exacerbation of alcoholic fatty liver disease (NAFLD) through SRSF6-associated RNA selective splicing mechanism, regulation of chronic lymphocytic leukemia and acute myeloid leukemia, and progression of colorectal cancer. This review focuses on the structure, function, and upstream pathways of DRAK2 and discusses the potential and challenges associated with the clinical application of DRAK2-based small-molecule inhibitors, with the aim of advancing DRAK2 research.
Collapse
Affiliation(s)
| | | | | | - Yong Wang
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
7
|
Fu H, Nicolet D, Mrózek K, Stone RM, Eisfeld A, Byrd JC, Archer KJ. Controlled variable selection in Weibull mixture cure models for high-dimensional data. Stat Med 2022; 41:4340-4366. [PMID: 35792553 PMCID: PMC9545322 DOI: 10.1002/sim.9513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 06/14/2022] [Accepted: 06/19/2022] [Indexed: 12/03/2022]
Abstract
Medical breakthroughs in recent years have led to cures for many diseases. The mixture cure model (MCM) is a type of survival model that is often used when a cured fraction exists. Many have sought to identify genomic features associated with a time-to-event outcome which requires variable selection strategies for high-dimensional spaces. Unfortunately, currently few variable selection methods exist for MCMs especially when there are more predictors than samples. This study develops high-dimensional penalized Weibull MCMs, which allow for identification of prognostic factors associated with both cure status and/or survival. We demonstrated how such models may be estimated using two different iterative algorithms. The model-X knockoffs method was combined with these algorithms to control the false discovery rate (FDR) in variable selection. Through extensive simulation studies, our penalized MCMs have been shown to outperform alternative methods on multiple metrics and achieve high statistical power with FDR being controlled. In an acute myeloid leukemia (AML) application with gene expression data, our proposed approach identified 14 genes associated with potential cure and 12 genes with time-to-relapse, which may help inform treatment decisions for AML patients.
Collapse
Affiliation(s)
- Han Fu
- Division of BiostatisticsCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
| | - Deedra Nicolet
- Clara D. Bloomfield Center for Leukemia Outcomes ResearchThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
- Alliance Statistics and Data Management CenterThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Krzysztof Mrózek
- Clara D. Bloomfield Center for Leukemia Outcomes ResearchThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - Richard M. Stone
- Dana‐Farber/Partners CancerHarvard UniversityBostonMassachusettsUSA
| | - Ann‐Kathrin Eisfeld
- Clara D. Bloomfield Center for Leukemia Outcomes ResearchThe Ohio State University Comprehensive Cancer CenterColumbusOhioUSA
| | - John C. Byrd
- Department of Internal MedicineUniversity of CincinnatiCincinnatiOhioUSA
| | - Kellie J. Archer
- Division of BiostatisticsCollege of Public Health, The Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
8
|
Shi X, Zhou Q, Huang B, Xia S, Jiang Y, Fang S, Lin J. Prognostic and immune-related value of STK17B in skin cutaneous melanoma. PLoS One 2022; 17:e0263311. [PMID: 35171924 PMCID: PMC8849620 DOI: 10.1371/journal.pone.0263311] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/16/2022] [Indexed: 11/26/2022] Open
Abstract
Skin cutaneous melanoma (SKCM) is a common cancer of which mortality is increasing continuously. Our study conducted a series of analyses on the clinical significance of Serine/threonine kinase 17B (STK17B) in SKCM to provide a new biomarker for diagnosis and treatment. The RNA-sequence data were obtained from The Cancer Genome Atlas and Genotype-Tissue Expression databases. The data of 468 SKCM patients were divided into STK17B high- and low-expression groups and analyzed by Bioconductor package to identify the differential expressed genes. The R package of "clusterProfiler" was used for Gene Ontology, Kyoto Encyclopedia of Genes and Genomes, and Gene-Set Enrichment Analysis analyses. A protein-protein interaction network and immune infiltration landscape were respectively constructed via STRING database and ssGSEA. STK17B had lower expression in SKCM than normal tissues. Besides, STK17B expression was significantly related to some clinicopathological characteristics in SKCM patients including T stage, Breslow depth, radiation therapy, melanoma Clark level, and pathologic stage. The Kaplan-Meier curve analyses revealed that the low expression of STK17B was correlated with poor overall survival and disease-specific survival. We constructed nomograms to predict the 1-, 3-, and 5-year survival of SKCM patients. The function enrichment analyses showed STK17B-related differential expressed genes were enriched in cellular differentiation and immune-related progress. STK17B expression level were positively correlated with infiltrating level of immune cells. In this study, we found that STK17B, which played an important role in immune infiltration, could be a new biomarker for diagnosis and prognosis in SKCM patients.
Collapse
Affiliation(s)
- Xueying Shi
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Qi Zhou
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Bingqian Huang
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, Liaoning, China
| | - Shilin Xia
- Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yuankuan Jiang
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Shifeng Fang
- Department of Ophthalmology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jingrong Lin
- Department of Dermatology, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
9
|
Desterke C, Bennaceur-Griscelli A, Turhan AG. EGR1 dysregulation defines an inflammatory and leukemic program in cell trajectory of human-aged hematopoietic stem cells (HSC). Stem Cell Res Ther 2021; 12:419. [PMID: 34294125 PMCID: PMC8296523 DOI: 10.1186/s13287-021-02498-0] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/08/2021] [Indexed: 12/24/2022] Open
Abstract
Background During aging, hematopoietic stem cells (HSC) lose progressively both their self-renewal and differentiation potential. The precise molecular mechanisms of this phenomenon are not well established. To uncover the molecular events underlying this event, we have performed a bioinformatics analysis of 650 single-cell transcriptomes. Methods Single-cell transcriptome analyses of expression heterogeneity, cell cycle, and cell trajectory in human cell compartment enriched in hematopoietic stem cell compartment were investigated in the bone marrow according to the age of the donors. Identification of aging-related nodules was identified by weighted correlation network analysis in this primitive compartment. Results The analysis of single-cell transcriptomes allowed to uncover a major upregulation of EGR1 in human-aged lineage−CD34+CD38− cells which present cell cycle dysregulation with reduction of G2/M phase according to less expression of CCND2 during S phase. EGR1 upregulation in aging hematopoietic stem cells was found to be independent of cell cycle phases and gender. EGR1 expression trajectory in aged HSC highlighted a signature enriched in hematopoietic and immune disorders with the best induction of AP-1 complex and quiescence regulators such as EGR1, BTG2, JUNB, and NR41A. Sonic Hedgehog-related TMEM107 transmembrane molecule followed also EGR1 cell trajectory. EGR1-dependent gene weighted network analysis in human HSC-associated IER2 target protein-specific regulators of PP2A activity, IL1B, TNFSF10 ligands, and CD69, SELP membrane molecules in old HSC module with immune and leukemogenic signature. In contrast, for young HSC which were found with different cell cycle phase progression, its specific module highlighted upregulation of HIF1A hypoxic factor, PDE4B immune marker, DRAK2 (STK17B) T cell apoptosis regulator, and MYADM myeloid-associated marker. Conclusion EGR1 was found to be connected to the aging of human HSC and highlighted a specific cell trajectory contributing to the dysregulation of an inflammatory and leukemia-related transcriptional program in aged human HSCs. EGR1 and its program were found to be connected to the aging of human HSC with dissociation of quiescence property and cell cycle phase progression in this primitive hematopoietic compartment. Supplementary Information The online version contains supplementary material available at 10.1186/s13287-021-02498-0.
Collapse
Affiliation(s)
| | - Annelise Bennaceur-Griscelli
- INSERM UA9, University Paris-Saclay, 94800, Villejuif, France.,ESTeam Paris Sud, INGESTEM National IPSC Infrastructure, University Paris-Saclay, 94800, Villejuif, France.,Division of Hematology, APHP-Paris Saclay University Hospitals, Le Kremlin Bicêtre 94275, 94800, Villejuif, France.,Faculty of Medicine, University Paris Saclay, 94275, Le Kremlin Bicêtre, France
| | - Ali G Turhan
- INSERM UA9, University Paris-Saclay, 94800, Villejuif, France. .,ESTeam Paris Sud, INGESTEM National IPSC Infrastructure, University Paris-Saclay, 94800, Villejuif, France. .,Division of Hematology, APHP-Paris Saclay University Hospitals, Le Kremlin Bicêtre 94275, 94800, Villejuif, France. .,Faculty of Medicine, University Paris Saclay, 94275, Le Kremlin Bicêtre, France.
| |
Collapse
|
10
|
Jiang L, Zhou J, Zhao S, Wang X, Chen Y. STK17B promotes the progression of ovarian cancer. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:475. [PMID: 33850872 PMCID: PMC8039663 DOI: 10.21037/atm-21-601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background Protein kinase is increasingly receiving widespread attention because of its role in the tumor progression. Serine/threonine protein kinase (STK) is an important family involved in the development of a variety of cancers. Many studies have shown that serine/threonine kinase 17B (STK17B) is highly expressed in a variety of malignant tumors and participate in proliferation and metastasis. However, the exact function of STK17B remains uncertain in ovarian cancer. Our study aims to investigate whether STK17B plays a role in the occurrence and development of epithelial ovarian cancer. Methods We employed quantitative reverse transcription polymerase chain reaction to detect the relative expression of STK17B in ovarian cancer tissues. STK17B was down-regulated and up-regulated in ovarian cancer cell lines by small interfering RNA and overexpressed plasmid, respectively. The effects of STK17B on proliferation, invasion and migration of ovarian cancer cells in vitro were analyzed by CCK-8 test, Transwell test, scratch test and EDU test. The tumorigenicity of subcutaneous xenograft tumor in nude mice to study the role of STK17B in tumorigenesis in vivo. Western Blotting analysis revealed that STK17B and EMT. Results STK17B expression was significantly increased in ovarian cancer tissues. The STK17B silencing suppressed cell progression, while the overexpression of STK17B promoted progression in vivo or in vitro. Western bolt showed that STK17B increased the invasion and migration of ovarian cancer cell by promoting the EMT process. Conclusions STK17B was highly expressed in epithelial ovarian cancer tissues and increased the proliferation, invasion and migration of ovarian cancer cells by promoting EMT process.
Collapse
Affiliation(s)
- Liping Jiang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Gynecology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Jinhua Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Shaojie Zhao
- Department of Gynecology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Xuzhen Wang
- Department of Breast Surgery, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi, China
| | - Youguo Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
11
|
Hou X, Li JY, Zhao M, Dai C, Li Y, Liu Y. Synthesis, Characterization, and DRAK2 Inhibitory Activities of Hydroxyaurone Derivatives. HETEROCYCLES 2021. [DOI: 10.3987/com-21-14481] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
12
|
Szoltysek K, Ciardullo C, Zhou P, Walaszczyk A, Willmore E, Rand V, Marshall S, Hall A, J. Harrison C, Eswaran J, Soundararajan M. DAP Kinase-Related Apoptosis-Inducing Protein Kinase 2 (DRAK2) Is a Key Regulator and Molecular Marker in Chronic Lymphocytic Leukemia. Int J Mol Sci 2020; 21:ijms21207663. [PMID: 33081245 PMCID: PMC7593912 DOI: 10.3390/ijms21207663] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/13/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023] Open
Abstract
Chronic lymphocytic leukemia (CLL) is the most common adult leukemia in the Western World and it is characterized by a marked degree of clinical heterogeneity. An impaired balance between pro- and anti-apoptotic stimuli determines chemorefractoriness and outcome. The low proliferation rate of CLL cells indicates that one of the primary mechanisms involved in disease development may be an apoptotic failure. Here, we study the clinical and functional significance of DRAK2, a novel stress response kinase that plays a critical role in apoptosis, T-cell biology, and B-cell activation in CLL. We have analyzed CLL patient samples and showed that low expression levels of DRAK2 were significantly associated with unfavorable outcome in our CLL cohort. DRAK2 expression levels showed a positive correlation with the expression of DAPK1, and TGFBR1. Consistent with clinical data, the downregulation of DRAK2 in MEC-1 CLL cells strongly increased cell viability and proliferation. Further, our transcriptome data from MEC-1 cells highlighted MAPK, NF-κB, and Akt and as critical signaling hubs upon DRAK2 knockdown. Taken together, our results indicate DRAK2 as a novel marker of CLL survival that plays key regulatory roles in CLL prognosis.
Collapse
MESH Headings
- Aged
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Cell Proliferation
- Cell Survival
- Death-Associated Protein Kinases/genetics
- Death-Associated Protein Kinases/metabolism
- Down-Regulation
- Female
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- MAP Kinase Signaling System
- Male
- Middle Aged
- NF-kappa B/genetics
- NF-kappa B/metabolism
- Protein Serine-Threonine Kinases/genetics
- Protein Serine-Threonine Kinases/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- Receptor, Transforming Growth Factor-beta Type I/genetics
- Receptor, Transforming Growth Factor-beta Type I/metabolism
Collapse
Affiliation(s)
- Katarzyna Szoltysek
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (K.S.); (C.C.); (E.W.); (A.H.); (C.J.H.)
- Maria Skłodowska-Curie Institute, Oncology Center, Gliwice Branch, 02-034 Gliwice, Poland
| | - Carmela Ciardullo
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (K.S.); (C.C.); (E.W.); (A.H.); (C.J.H.)
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
| | - Peixun Zhou
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3JN, UK; (P.Z.); (V.R.)
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Anna Walaszczyk
- Institute of Biosciences, International Centre for Life, Newcastle University, Newcastle upon Tyne NE1 7RU, UK;
| | - Elaine Willmore
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (K.S.); (C.C.); (E.W.); (A.H.); (C.J.H.)
| | - Vikki Rand
- School of Health & Life Sciences, Teesside University, Middlesbrough TS1 3JN, UK; (P.Z.); (V.R.)
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
| | - Scott Marshall
- Department of Haematology, City Hospitals Sunderland NHS Trust, Sunderland SR4 7TP, UK;
| | - Andy Hall
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (K.S.); (C.C.); (E.W.); (A.H.); (C.J.H.)
| | - Christine J. Harrison
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (K.S.); (C.C.); (E.W.); (A.H.); (C.J.H.)
| | - Jeyanthy Eswaran
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE1 7RU, UK; (K.S.); (C.C.); (E.W.); (A.H.); (C.J.H.)
- Newcastle University Medicine Malaysia (NUMed Malaysia), EduCity, Iskandar 79200, Johor, Malaysia
- Correspondence: or (J.E); (M.S.)
| | - Meera Soundararajan
- Department of Applied Sciences, Faculty of Health and Life Sciences, Northumbria University, Newcastle upon Tyne NE1 8ST, UK
- Correspondence: or (J.E); (M.S.)
| |
Collapse
|
13
|
Lan Y, Han J, Wang Y, Wang J, Yang G, Li K, Song R, Zheng T, Liang Y, Pan S, Liu X, Zhu M, Liu Y, Meng F, Mohsin M, Cui Y, Zhang B, Subash S, Liu L. STK17B promotes carcinogenesis and metastasis via AKT/GSK-3β/Snail signaling in hepatocellular carcinoma. Cell Death Dis 2018; 9:236. [PMID: 29445189 PMCID: PMC5833726 DOI: 10.1038/s41419-018-0262-1] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 11/10/2017] [Accepted: 11/24/2017] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) is a lethal malignancy worldwide with frequent intrahepatic and distant metastasis. Elucidating the underlying molecular mechanism that modulates HCC progression is critical for exploring novel therapeutic strategies. Serine/Threonine Kinase 17B (STK17B) is upregulated in HCC tissues, but its role in HCC progression remains elusive. In the present studies, we reported that STK17B had a critical role in HCC progression. STK17B was significantly upregulated in HCC cell lines and specimens, and patients with ectopic STK17B expression characterized with poor clinicopathological features. In vitro and in vivo assay demonstrated that inhibition of STK17B markedly inhibits HCC tumorigenesis and metastasis, while STK17B overexpression promoted these processes. Furthermore, we found that STK17B promoted EMT process via activating AKT/GSK-3β/Snail signal pathway, and miR-455-3p was identified as the upstream regulator of STK17B. Combination of high level of STK17B and low level of miR-455-3p predicted poor prognosis with higher accuracy for HCC patients. In conclusion, our research demonstrated that STK17B promotes HCC progression, induces EMT process via activating AKT/GSK-3β/Snail signal and predicts poor prognosis in HCC.
Collapse
Affiliation(s)
- Yaliang Lan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Jihua Han
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yan Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Jiabei Wang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Guangchao Yang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Keyu Li
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Ruipeng Song
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Tongsen Zheng
- Department of Gastrointestinal Medical Oncology, The Affiliated Tumor Hospital of Harbin Medical University, Harbin, China
| | - Yingjian Liang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Shangha Pan
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Xirui Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Mingxi Zhu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yao Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Fanzheng Meng
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Manzoor Mohsin
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Yifeng Cui
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Bo Zhang
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Sharma Subash
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China
| | - Lianxin Liu
- Department of Hepatic Surgery, The First Affiliated Hospital of Harbin Medical University, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, Harbin, China.
| |
Collapse
|
14
|
Synergistic cooperation between ABT-263 and MEK1/2 inhibitor: effect on apoptosis and proliferation of acute myeloid leukemia cells. Oncotarget 2016; 7:845-59. [PMID: 26625317 PMCID: PMC4808037 DOI: 10.18632/oncotarget.6417] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2015] [Accepted: 11/16/2015] [Indexed: 12/14/2022] Open
Abstract
In spite of intensive research to improve treatment of acute myeloid leukemia (AML) more than half of all patients continue to develop a refractory disease. Therefore there is need to improve AML treatment. The overexpression of the BCL-2 family anti-apoptotic members, like BCL-2 or BCL-xL has been largely reported in lymphoid tumors but also in AML and other tumors. To counteract the anti-apoptotic effect of BCL-2, BH3 mimetics have been developed to target cancer cells. An increase in activity of ERK1/2 mitogen activated protein (MAP) kinase has also been reported in AML and might be targeted by MEK1/2 inhibitors. Hence, in the current work, we investigated whether the association of a BH3 mimetic such ABT-263 and the MEK1/2 inhibitor pimasertib (MEKI), was efficient to target AML cells. A synergistic increasing of apoptosis was observed in AML cell lines and in primary cells without affecting normal bone marrow cells. Such cooperation was confirmed on tumor growth in a mouse xenograft model of AML. In addition we demonstrated that MEKI sensitized the cells to apoptosis through its ability to promote a G1 cell cycle arrest. So, this combination of a MAP Kinase pathway inhibitor and a BH3 mimetic could be a promising strategy to improve the treatment of AML.
Collapse
|
15
|
Overexpression of c-Myb is associated with suppression of distant metastases in colorectal carcinoma. Tumour Biol 2016; 37:10723-9. [DOI: 10.1007/s13277-016-4956-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Accepted: 02/02/2016] [Indexed: 01/09/2023] Open
|
16
|
Abstract
Strict control of tissue-specific gene expression plays a pivotal role during lineage commitment. The transcription factor c-Myb has an essential role in adult haematopoiesis and functions as an oncogene when rearranged in human cancers. Here we have exploited digital genomic footprinting analysis to obtain a global picture of c-Myb occupancy in the genome of six different haematopoietic cell-types. We have biologically validated several c-Myb footprints using c-Myb knockdown data, reporter assays and DamID analysis. We show that our predicted conserved c-Myb footprints are highly dependent on the haematopoietic cell type, but that there is a group of gene targets common to all cell-types analysed. Furthermore, we find that c-Myb footprints co-localise with active histone mark H3K4me3 and are significantly enriched at exons. We analysed co-localisation of c-Myb footprints with 104 chromatin regulatory factors in K562 cells, and identified nine proteins that are enriched together with c-Myb footprints on genes positively regulated by c-Myb and one protein enriched on negatively regulated genes. Our data suggest that c-Myb is a transcription factor with multifaceted target regulation depending on cell type.
Collapse
|
17
|
Harris TL, McGargill MA. Drak2 Does Not Regulate TGF-β Signaling in T Cells. PLoS One 2015; 10:e0123650. [PMID: 25951457 PMCID: PMC4423867 DOI: 10.1371/journal.pone.0123650] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 02/10/2015] [Indexed: 12/31/2022] Open
Abstract
Drak2 is a serine/threonine kinase expressed highest in T cells and B cells. Drak2-/- mice are resistant to autoimmunity in mouse models of type 1 diabetes and multiple sclerosis. Resistance to these diseases occurs, in part, because Drak2 is required for the survival of autoreactive T cells that induce disease. However, the molecular mechanisms by which Drak2 affects T cell survival and autoimmunity are not known. A recent report demonstrated that Drak2 negatively regulated transforming growth factor-β (TGF-β) signaling in tumor cell lines. Thus, increased TGF-β signaling in the absence of Drak2 may contribute to the resistance to autoimmunity in Drak2-/- mice. Therefore, we examined if Drak2 functioned as a negative regulator of TGF-β signaling in T cells, and whether the enhanced susceptibility to death of Drak2-/- T cells was due to augmented TGF-β signaling. Using several in vitro assays to test TGF-β signaling and T cell function, we found that activation of Smad2 and Smad3, which are downstream of the TGF-β receptor, was similar between wildtype and Drak2-/- T cells. Furthermore, TGF-β-mediated effects on naïve T cell proliferation, activated CD8+ T cell survival, and regulatory T cell induction was similar between wildtype and Drak2-/- T cells. Finally, the increased susceptibility to death in the absence of Drak2 was not due to enhanced TGF-β signaling. Together, these data suggest that Drak2 does not function as a negative regulator of TGF-β signaling in primary T cells stimulated in vitro. It is important to investigate and discern potential molecular mechanisms by which Drak2 functions in order to better understand the etiology of autoimmune diseases, as well as to validate the use of Drak2 as a target for therapeutic treatment of these diseases.
Collapse
Affiliation(s)
- Tarsha L. Harris
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
| | - Maureen A. McGargill
- Department of Immunology, St. Jude Children’s Research Hospital, Memphis, Tennessee, United States of America
- Integrated Biomedical Sciences Program, University of Tennessee Health Science Center, Memphis, Tennessee, United States of America
- * E-mail:
| |
Collapse
|
18
|
Edwards BA, Harris TL, Floersh H, Lukens JR, Zaki MH, Vogel P, Kanneganti TD, Bui JD, McGargill MA. Drak2 is not required for tumor surveillance and suppression. Int Immunol 2015; 27:161-6. [PMID: 25568303 DOI: 10.1093/intimm/dxu146] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Drak2 is a promising therapeutic target to treat organ-specific autoimmune diseases such as type 1 diabetes and multiple sclerosis without causing generalized immune suppression. Inhibition of Drak2 may also prevent graft rejection following organ transplantation. However, Drak2 may function as a critical tumor suppressor, which would challenge the prospect of targeting Drak2 for therapeutic treatment. Thus, we examined the susceptibility of Drak2 (-/-) mice in several tumor models. We show that Drak2 is not required to prevent tumor formation in a variety of settings. Therefore, Drak2 does not function as an essential tumor suppressor in in vivo tumor models. These data further validate Drak2 as a viable therapeutic target to treat autoimmune disease and graft rejection. Importantly, these data also indicate that while Drak2 may induce apoptosis when overexpressed in cell lines, it is not an essential tumor suppressor.
Collapse
Affiliation(s)
- Benjamin A Edwards
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| | - Tarsha L Harris
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| | - Helen Floersh
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| | - John R Lukens
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| | - Md Hasan Zaki
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| | - Peter Vogel
- Department of Veterinary Pathology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| | - Jack D Bui
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Maureen A McGargill
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, MS 351, Memphis, TN 38105, USA
| |
Collapse
|
19
|
Ye P, Zhao L, McGirr C, Gonda TJ. MYB down-regulation enhances sensitivity of U937 myeloid leukemia cells to the histone deacetylase inhibitor LBH589 in vitro and in vivo. Cancer Lett 2014; 343:98-106. [PMID: 24075958 DOI: 10.1016/j.canlet.2013.09.022] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2013] [Revised: 09/16/2013] [Accepted: 09/18/2013] [Indexed: 11/30/2022]
Abstract
The effect of combining MYB suppression with the histone deacetylase inhibitor LBH589 was studied in human myeloid leukemia cell lines. MYB knockdown inhibited proliferation and induced apoptosis in U937 and K562 cells in vitro, and also sensitized both to the pro-apoptotic effect of LBH589. This was accompanied by enhanced expression of the pro-apoptotic BCL2 family members BOK and BIM. U937 cells carrying inducible MYB shRNA were also transplanted into NOD/SCID mice. The combination of MYB knockdown and LBH589 prolonged survival compared to either treatment alone, suggesting that further development of such combinations might lead to effective and safe leukemia therapies.
Collapse
Affiliation(s)
- Ping Ye
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4102, Australia; The University of Queensland Diamantina Institute, Brisbane, Queensland 4102, Australia
| | - Liang Zhao
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Crystal McGirr
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4102, Australia; The University of Queensland Diamantina Institute, Brisbane, Queensland 4102, Australia
| | - Thomas J Gonda
- School of Pharmacy, The University of Queensland, Brisbane, Queensland 4102, Australia; The University of Queensland Diamantina Institute, Brisbane, Queensland 4102, Australia.
| |
Collapse
|
20
|
Zhao L, Ye P, Gonda TJ. The MYB proto-oncogene suppresses monocytic differentiation of acute myeloid leukemia cells via transcriptional activation of its target gene GFI1. Oncogene 2013; 33:4442-9. [PMID: 24121275 DOI: 10.1038/onc.2013.419] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Revised: 08/14/2013] [Accepted: 09/02/2013] [Indexed: 12/20/2022]
Abstract
The MYB gene is a master regulator of hematopoiesis and contributes to leukemogenesis in several species including humans. Although it is clear that MYB can promote proliferation, suppress apoptosis and block differentiation, the identities of the MYB target genes that mediate these effects have only been partially elucidated. Several studies, including our own, have collectively identified substantial numbers of MYB target genes, including candidates for each of these activities; however, functional validation, particularly in the case of differentiation suppression, has lagged well behind. Here we show that GFI1, which encodes an important regulator of hematopoietic stem cell (HSC) function and granulocytic differentiation, is a direct target of MYB in myeloid leukemia cells. Chromatin immunoprecipitation and reporter studies identified a functional MYB-binding site in the promoter region of GFI, whereas ectopic expression and small hairpin RNA-mediated knockdown of MYB resulted in concomitant increases and decreases, respectively, in GFI1 expression. We also demonstrate that GFI1, like MYB, can block the induced monocytic differentiation of a human acute myeloid leukemia cell line, and most importantly, that GFI1 is essential for MYB's ability to block monocytic differentiation. Thus, we have identified a target of MYB that is a likely mediator of its myeloid differentiation-blocking activity, and which may also be involved in MYB's activities in regulating normal HSC function and myeloid differentiation.
Collapse
Affiliation(s)
- L Zhao
- The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia
| | - P Ye
- 1] The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia [2] School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Brisbane, Queensland, Australia
| | - T J Gonda
- 1] The University of Queensland Diamantina Institute, Brisbane, Queensland, Australia [2] School of Pharmacy, The University of Queensland, Pharmacy Australia Centre of Excellence, Brisbane, Queensland, Australia
| |
Collapse
|